Abstract
Background
PI3K/mTOR inhibition leads to apoptosis of NOTCH1-mutant head and neck squamous cell carcinoma (HNSCC) cells. We tested the efficacy of the PI3K/mTOR inhibitor bimiralisib in patients with NOTCH1-mutant HNSCC.
Methods
Patients with recurrent/metastatic NOTCH1-mutant HNSCC who had progressed during chemotherapy and immunotherapy received bimiralisib until unacceptable toxicity or progression. To assess whether NOTCH1 mutations can be detected in blood, we measured circulating tumor DNA (ctDNA). To assess activated NOTCH1 protein levels, we quantitated cleaved NOTCH1 (cl-NOTCH) by immunohistochemistry.
Results
Eight patients were treated, and 6 were evaluable for response. The objective response rate was 17%. For all 8 patients, median progression-free and overall survival was 5 and 7 months, respectively. Bimiralisib was well tolerated, with expected hyperglycemia. Pharmacokinetic values were consistent with published studies. NOTCH1 mutations were detected in 83.3% of ctDNA. Staining for tumor cl-NOTCH1 was negative. The trial closed early due to sponsor insolvency.
Conclusion
Although the trial was small, outcomes with bimiralisib were better than the historical standard of care; Results will need to be confirmed in a larger trial. The lack of cl-NOTCH1 was consistent with loss-of-function mutations and validated our mutation function algorithm. The ability to detect NOTCH1 mutations in blood will help future studies. (ClinicalTrials.gov Identifier: NCT03740100).
Keywords: head and neck squamous cell carcinoma, PI3 kinase, mTOR, NOTCH1, pharmacokinetics
Recurrent head and neck squamous cell carcinoma (HNSCC) remains lethal despite recent advances with immunotherapy, and effective targeted therapies are still needed. This article reports on the efficacy of the PI3K/mTOR inhibitor bimiralisib in patients with NOTCH1-mutant HNSCC.
Lessons Learned.
In a small cohort of patients with chemotherapy- and immunotherapy-refractory recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), both the responses and the prolonged stable disease following single-agent bimiralisib appeared to be promising.
Because NOTCH1 loss-of-function mutations are common in other squamous cell carcinomas (esophagus, lung, and skin), these findings may translate beyond HNSCC.
The lack of cleaved NOTCH1 expression in tumors validated the authors’ algorithm for determining NOTCH1 mutation function.
The ability to detect NOTCH1 mutations in blood will help future studies.
Discussion
The identification of genomic alterations in HNSCC has yet to improve patient outcomes because most alterations are in tumor suppressors, including NOTCH1, which is mutated in ~20% of patients. Recurrent HNSCC remains lethal despite recent advances with immunotherapy, and effective targeted therapies are still needed. To address this need, we showed that PI3K/mTOR inhibition induced apoptosis in HNSCC cell lines with NOTCH1 loss-of-function (LOF) mutations in vitro and in vivo. In a phase I study of single-agent bimiralisib, one patient with HNSCC with a NOTCH1-mutation was treated and experienced a partial response that lasted for 9 months. In the current study, we tested the hypothesis that the pan-class I PI3K/mTOR antagonist bimiralisib would result in cancer cell apoptosis and concomitant tumor shrinkage in NOTCH1-mutant HNSCC. To determine if NOTCH1 mutations can be detected in blood and monitor response to therapy, we assessed ctDNA at baseline, at week 7, and at progression. We evaluated the functional impact of NOTCH1 mutations using our novel algorithm. To determine if tumors retained any NOTCH1 protein activity, we assessed the presence of activated NOTCH1 in pretreatment tumors using an antibody that detects activated NOTCH1 (cl-NOTCH1).
In the 6 evaluable patients with recurrent or metastatic (R/M) HNSCC that was chemotherapy and immunotherapy resistant, bimiralisib had an objective response rate of 17%. For all 8 treated patients, the median OS and PFS was 7 and 5 months, respectively. One patient had a confirmed partial response with a 49% reduction in the target lesion size (Fig. 1A-D). She remained in the study until day 157 when the protocol was terminated. Three patients had confirmed stable disease before disease progression on days 91, 138, or 167. An additional patient was noted to have clear shrinkage on imaging. Tumor dimensions decreased from 19 mm × 16 mm to 17 mm × 8 mm on day 42 (Fig. 1E, F), but this response was technically stable disease according to RECIST. One patient had progressive disease on day 83. Two patients were not evaluable for response based on toxicity from cancer progression on day 8 and day 9.
This is the only clinical trial to test the therapeutic vulnerability of NOTCH1-mutant HNSCC to any class of drugs. Future studies in NOTCH1-mutant cancers will be facilitated by the validation of our algorithm to predict NOTCH1 mutation function and the ability to detect NOTCH1 mutations in blood. Although the trial was small, the outcomes were promising in a group of HNSCC patients with a very poor prognosis. Because NOTCH1 loss-of-function mutations are common in other squamous cell carcinomas, including those of the esophagus and lung, our findings may translate beyond HNSCC.
Trial Information
Disease | Head and neck squamous cell carcinoma (HNSCC) |
Stage of disease/treatment | Recurrent or metastatic |
Prior therapy | Cisplatin-based chenotherapy. Anti-PD1 mmunotherapy |
Type of study | Phase II, nonrandomized, non-blinded, single-arm study |
Primary endpoint | The objective response rate, including confirmed complete response and confirmed partial response, according to RECIST version 1.1 |
Secondary endpoints | Safety, progression-free survival, overall survival, pharmacokinetic analysis, and biomarkers (ctDNA, cl-NOTCH) |
Investigator’s analysis | Active and should be pursued further |
Additional Details of Endpoints or Study Design
Patient Selection
Adults with an Eastern Cooperative Oncology Group performance status of ≤2, measurable disease according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1,1 and histologically confirmed recurrent or metastatic (R/M) HNSCC for which no standard curative or life-prolonging therapy was available were eligible. All patients had already received platinum chemotherapy and α-PD1 immunotherapy unless unable to tolerate them.
The presence of a NOTCH1 mutation in the HNSCC tissue had to be determined by Clinical Laboratory Improvement Amendments–certified sequencing results. We excluded NOTCH1 splice mutations in exons 33 or 34; truncating mutations in carboxy-terminal regions associated with activation, including the transcription activation domain and proline-, glutamate-, serine-, and threonine-rich domains; and non-truncating mutations in the negative regulatory region and heterodimerization domains. Patients with NOTCH1 mutations in all other regions were further evaluated for eligibility using an algorithm as described2 which predicts the likelihood of inactivation based on both the specific NOTCH1 protein domain affected and the PROVEAN/SIFT3,4 scores of amino acid changes.
We defined adequate bone marrow, liver, and renal functions as follows: platelet count ≥100 × 109/L, absolute neutrophil count ≥1.5 × 109/L, hemoglobin ≥9 g/dL, alanine aminotransferase and aspartate aminotransferase ≤2.5 × upper limit of normal, serum total bilirubin ≤upper limit of normal, glomerular filtration rate ≥30 mL/minute (Cockcroft-Gault), and fasting plasma glucose ≤150 mg/dL.
Study Design
All patients received 140 mg bimiralisib (PQR309) orally once daily on 2 consecutive days followed by 5 days without treatment weekly until unacceptable toxicity, tumor progression, patient request for withdrawal, investigator judgment, or death. The primary endpoint was to determine the objective response rate, including confirmed complete response and confirmed partial response according to RECIST version 1.1.1 Secondary and exploratory endpoints included safety, progression-free survival (PFS), overall survival, pharmacokinetic (PK) analysis, and biomarkers described below.
To minimize accrual if bimiralisib was ineffective, we used a Simon optimal 2-stage design. In order to have 80% power to detect a response rate of 30% (one-sided α = 0.05 and β = 0.20), we planned to enroll up to 10 patients in the first stage. If ≥2 patients had an objective response, then we had planned to enroll an additional 19 patients in the second stage. Radiologic tumor assessments were performed using computed tomography at baseline and every 6 weeks. Safety was continuously monitored.
The trial was registered on ClinicalTrials.gov (NCT03740100). All patients provided written, informed consent. The study was performed in accordance with the ethical principles of the declaration of Helsinki and in agreement with the International Council for Harmonization guidelines on Good Clinical Practice E6 R2. One patient (patient 9) transitioned to an expanded access program to receive bimiralisib when NCT03740100 closed and the patient received identical treatment and assessments as in NCT03740100 until the expanded access program closed.
Statistical Analysis
Demographic and clinical characteristics were summarized using univariate statistics, including mean, median, and standard deviation or frequency and percentage. Objective response rate was summarized by frequency and percentage with a 2-sided 95% Agresti-Coull CI.5 Duration of response, PFS, and overall survival were analyzed using the Kaplan-Meier method.6 All statistical analyses were performed using R statistical software (version 3.6.3, R Core Team, 2020). Survival modeling was performed using the “survival” package.7
Time to response, duration of response, PFS, and overall survival were analyzed using the Kaplan-Meier method.6 Time to response was defined as the time from first study drug administration to the first documentation of response (complete or partial). Duration of response was defined as the time from the date of the first confirmed response to the first documentation of disease progression. PFS was defined as the time from first study drug administration to disease progression or death due to any cause. Overall survival was defined as the time from first study drug administration to death due to any cause.
PK Analysis
Blood was collected before oral dose administration and 1 hour after oral dose administration on days 1, 8, 22, 43, and 64 after the start of therapy. Bimiralisib levels were determined as previously described using high-performance liquid chromatography8-11 by Charles River Laboratories (Ashland, Ohio). The pre-dose levels were the trough levels and post-dose levels were the assumed peak serum concentration (Cmax) values. Cmax and trough accumulation ratios were calculated by dividing each patient’s treatment day concentration by their initial available drug concentration—day 1 for Cmax and day 8 for trough. Statistical differences among groups were identified by analysis of variance using Statistica version 13.1 (TIBCO Software Inc., Palo Alto, California); P < .05 was considered statistically significant
Cell-Free DNA
Peripheral blood samples were collected in K3 EDTA tubes from patients who consented to the optional blood collection protocol. Samples were processed within 30 minutes of collection. Plasma was obtained through centrifugation at 1500-2000 g for 10 minutes at 4 °C. Plasma was stored at 80 °C until further processing. Cell-free DNA was isolated using QIAamp Circulating Nucleic Acid Kit (QIAGEN, Hilden, Germany) and was initially quantified using Quant-iT Picogreen dsDNA Assay Kit (Thermofisher Scientific, Massachusetts) on SpectraMax M2 (Molecular Devices, California). Whenever feasible, 30 ng of DNA was used to prepare libraries that were sequenced on NovaSeq 6000 at The University of Texas MD Anderson Cancer Center Advanced Technology Genomics Core Laboratory using TruSight Oncology 500 ctDNA kit (TSO500; Illumina, California). TSO500 is a targeted next-generation sequencing assay that can sequence 523 genes and detect different DNA variants, including exon coverage. Bioinformatics analysis was done using DRAGEN analysis software and DRAGEN server version 3 (Illumina). We assessed the detection rate and variant allele frequency (VAF) of NOTCH1 mutations that were detected in tissue. For further analysis, the obtained mutations were filtered to exclude synonymous mutations, germline mutations, mutations that are not annotated in the Catalogue of Somatic Mutations in Cancer database, and mutations with FATHMM prediction as non-pathogenic.
Immunohistochemistry (IHC) for NOTCH1
To assess activated NOTCH1 protein levels in archival tumor material, we quantitated cleaved NOTCH1 (cl-NOTCH1, NICD) using an anti-cl-NOTCH1 antibody that was previously validated12-14 using IHC. The anti-cl-NOTCH1 antibody from Cell Signaling Technology (Cat # 4147) was previously validated to work in IHC to detect cl-NOTCH1 in formalin-fixed, paraffin-embedded sections. The anti-cl-NOTCH1 antibody recognizes an epitope on the intracellular NOTCH1 fragment that becomes accessible only when NOTCH1 is cleaved between Gly1743 and Val1744 in the transmembrane domain, which happens only during NOTCH1 activation or signaling.
Formalin-fixed, paraffin-embedded slides from patients or control cell pellets were dewaxed, treated with a 3:1 ratio of EZ antigen retrieval buffer EZ AR10 (pH 10) to EZ AR2 (pH 8.5), microwaved for 10 minutes at 98 °C, rinsed in deionized H2O and phosphate-buffered saline, and loaded onto an IntelliPATH autostainer for detection with a rabbit monoclonal antibody to cl-Notch1. Briefly, slides were washed in BOND buffer (Leica Biosystems), incubated with Leica endogenous peroxide blocker for 5 minutes, rinsed, incubated for 30 minutes with anti-cl-NOTCH1 diluted 1:100 in antibody dilution buffer (Cell Signaling Technology, #12378), washed, detected with Leica BOND Polymer anti-rabbit horseradish peroxidase for 8 minutes, rinsed, and developed with BioGenex DAB (prediluted at 1 drop per milliliter) for 5 minutes before staining with hematoxylin. All autostainer incubations were at room temperature. A positive control cell line for IHC was created by first knocking out wt NOTCH1 in the HNSCC cell line FADU with CRISPR and then expressing a cDNA corresponding to the cleaved/activated intracellular NOTCH1 molecule that was driven by a doxycycline-inducible promoter. We had planned to use a semi-quantitative/ordered categorical expression scoring system for IHC.12-14
Drug Information
Generic/working name | Bimiralisib |
Company name | PIQUR Therapeutics AG (Basel, Switzerland) |
Drug type | Small molecule |
Drug class | PI3K/mTOR |
Dose | 140 |
Unit | Mg |
Route | Oral |
Schedule of administration | Daily on 2 consecutive days weekly |
Patient Characteristics
Number of patients, male | 6 |
Number of patients, female | 2 |
Stage | IV |
Age: Median (range) | 65.5 (50-71) years |
Number of prior systemic therapies: median(range) | 3 (2-7) |
Performance status: ECOG | 0: 4 1: 4 2: 0 3: 0 4: 0 |
Cancer types or histologic subtypes | HNSCC, Oral cavity squamous carcinoma (SCC), human papillomavirus (HPV)-negative: 4; HNSCC, Oropharynx SCC, HPV-positive, 3; HNSCC, Oropharynx SCC, HPV unknown,1. |
Primary Assessment Method
Title | Objective response rate |
Number of patients screened | 10 |
Number of patients enrolled | 8 |
Number of patients evaluable for toxicity | 8 |
Number of patients evaluated for efficacy | 6 |
Evaluation method | RECIST 1.1 |
Response assessment, CR | 0 (0%) |
Response assessment, PR | 1 (17%) |
Response assessment, SD | 4 (67%) |
Duration assessments, median, PFS | 147 days (CI: 39-199) |
Duration assessments, median, TTP | 167 days (CI: 83-inf) |
Duration assessments, median, OS | 215 days (CI: 39-inf) |
Response duration | 111 days |
Duration of treatment | 87 days |
Outcome Notes
Between January 2019 and August 2020, 10 patients consented to receive the study treatment at MD Anderson; however, 2 of these patients died of cancer-related complications prior to treatment initiation, so 8 were treated. Per study design, all patients had recurrent or metastatic HNSCC harboring NOTCH1 mutations predicted to be loss of function (LOF). All had been treated with prior immuno- and chemotherapy with the exception of patient 4 who was unable to tolerate chemotherapy. The median number of prior systemic therapies for recurrent or metastatic HNSCC was 3. Most had also received prior definitive radiotherapy. The trial was closed early because the sponsoring company dissolved.
Overall, bimiralisib was well tolerated, with the expected toxicities (eg, hyperglycemia), cancer-related adverse events (AEs), and cancer progression accounting for all significant toxicity (Table 1). In total, 64 treatment-emergent AEs, defined as AEs that started or worsened during therapy, occurred during the study. All 8 patients experienced at least one treatment-emergent AE. No patients experienced an AE leading to dose reduction. AEs irrespective of causality occurring in ≥25% of patients included fatigue (3 patients, 37.5%), nausea (3 patients, 37.5%), vomiting (2 patients, 25.0%), hyperglycemia (3 patients, 37.5%), skin infection (2 patients, 25%), aspiration pneumonia (2 patients, 25.0%), rash (2 patients, 25.0%), and confusional state (2 patients, 25.0%).
Table 1.
Adverse event | Bimiralisib (n = 8), n (%) | Total number of events | ||||
---|---|---|---|---|---|---|
Grade 1 | Grade 2 | Grade 3/ 4 | Grade 1 | Grade 2 | Grade 3/ 4 | |
Any adverse event | 8 (100.0) | 3 (37.5) | 5 (62.5) | 29 | 13 | 15 |
General disorders and administration site conditions | 5 (62.5) | 1 (12.5) | 1 (12.5) | 6 | 1 | 1 |
Fatigue | 2 (25.0) | 1 (12.5) | 0 | 2 | 1 | 0 |
Asthenia | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Chest pain | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Disease progression | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Gait disturbance | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Pyrexia | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Gastrointestinal disorders | 2 (25.0) | 3 (37.5) | 0 | 2 | 5 | 0 |
Nausea | 1 (12.5) | 2 (25.0) | 0 | 1 | 2 | 0 |
Vomiting | 0 | 2 (25.0) | 0 | 0 | 2 | 0 |
Diarrhea | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Gastroesophageal reflux disease | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Infections and infestations | 0 | 2 (25.0) | 3 (37.5) | 0 | 3 | 3 |
Skin infection | 0 | 1 (12.5) | 1 (12.5) | 0 | 1 | 1 |
Cellulitis | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Osteomyelitis acute | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Sepsis | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Wound infection | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Metabolism and nutrition disorders | 1 (12.5) | 2 (25.0) | 2 (25.0) | 1 | 3 | 2 |
Hyperglycemia | 1 (12.5) | 0 | 2 (25.0) | 1 | 0 | 2 |
Decreased appetite | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Dehydration | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Hypocalcemia | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Investigations | 3 (37.5) | 0 | 1 (12.5) | 3 | 0 | 2 |
Alanine aminotransferase increased | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Aspartate aminotransferase increased | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Blood creatinine increased | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Blood glucose increased | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Platelet count decreased | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Respiratory, thoracic and mediastinal disorders | 1 (12.5) | 0 | 3 (37.5) | 1 | 0 | 4 |
Pneumonia aspiration | 0 | 0 | 2 (25.0) | 0 | 0 | 2 |
Acute respiratory distress syndrome | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Cough | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Laryngeal stenosis | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Skin and subcutaneous tissue disorders | 2 (25.0) | 0 | 0 | 4 | 0 | 0 |
Rash | 2 (25.0) | 0 | 0 | 2 | 0 | 0 |
Dry skin | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Pruritus | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Nervous system disorders | 1 (12.5) | 1 (12.5) | 0 | 2 | 1 | 0 |
Dysarthria | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Presyncope | 0 | 1 (12.5) | 0 | 0 | 1 | 0 |
Seizure | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Ear and labyrinth disorders | 2 (25.0) | 0 | 0 | 2 | 0 | 0 |
Ear pain | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Tinnitus | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Psychiatric disorders | 2 (25.0) | 0 | 0 | 2 | 0 | 0 |
Confusional state | 2 (25.0) | 0 | 0 | 2 | 0 | 0 |
Renal and urinary disorders | 1 (12.5) | 0 | 1 (12.5) | 1 | 0 | 1 |
Renal injury | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Urinary tract discomfort | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Vascular disorders | 1 (12.5) | 0 | 1 (12.5) | 1 | 0 | 1 |
Hemorrhage | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Hypotension | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Musculoskeletal and connective tissue disorders | 1 (12.5) | 0 | 0 | 2 | 0 | 0 |
Neck pain | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Pain in jaw | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Blood and lymphatic system disorders | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Anemia | 0 | 0 | 1 (12.5) | 0 | 0 | 1 |
Eye disorders | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Eye disorder | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Injury, poisoning and procedural complications | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Sunburn | 1 (12.5) | 0 | 0 | 1 | 0 | 0 |
Five patients (62.5%) experienced 14 severe AEs (SAEs, Common Terminology Criteria for Adverse Events grade ≥3). Two patients died during the study, one of local disease progression with associated infection and hemorrhage (patient 5) and one on day 8 of therapy from aspiration pneumonia (patient 7, details below), a common complication of recurrent HNSCC. Both events were unrelated to treatment with bimiralisib. During the on-treatment period, a further 10 nonfatal SAEs were reported that were not related to bimiralisib. Patient 3 chose to discontinue the study on day 9 after experiencing tumor-related laryngeal narrowing that required an emergency tracheostomy, making further travel to Houston problematic. Patient 9 had locally advanced disease with several cancer-related AEs during her 5 months participating in the study: aspiration pneumonia, local infection, and osteomyelitis.
Two SAEs—both hyperglycemia—were related to treatment with bimiralisib. Hyperglycemia is a well-known effect of PI3K inhibitors, including bimiralisib.15 One patient’s plasma glucose subsequently returned to near-normal values with medical management while the patient was participating in the study (patient 9). The other patient (patient 7) was hospitalized with grade 3 hyperglycemia that resolved, but he subsequently developed pneumonia, sepsis, respiratory distress, and acute renal failure due to aspiration from tumor-related dysphagia, which led to study discontinuation. Patient 4 discontinued participation in the study owing to grade 3 elevated liver function test results (alanine aminotransferase and aspartate aminotransferase) related to treatment with bimiralisib, and these levels normalized 28 days after therapy discontinuation. In total, 7 patients (87.5%) experienced 23 AEs (any severity) that were suspected to be related to treatment with bimiralisib.
One patient had a confirmed partial response with a 49% reduction in the target lesion size. She remained in the study until day 157 when the protocol was terminated; her final computed tomography scan during the study showed that she maintained this response, with a 45% reduction in the target lesion size compared with baseline. Three patients had confirmed stable disease before experiencing disease progression on days 91, 138, or 167. An additional patient with stable disease according to RECIST 1.1 had a notable decrease in tumor dimensions from 19 mm × 16 mm to 17 mm × 8 mm on day 42. She discontinued therapy owing to elevated liver function test results. One patient had progressive disease on day 83. Two patients were not evaluable based on toxicity described above. Among the 6 evaluable patients, the objective response rate was 17% (1/6), with 95% CI 1-58%. For all 8 patients, the median PFS was approximately 5 months (147 days, 95% CI 39-199 days) and the median overall survival was 7 months (215 days, 95% CI 39 days-infinity). In the 6 evaluable patients, the duration of response was 111 days (Figure 2).
To assess whether NOTCH1 mutations can be detected in baseline ctDNA samples and to explore potential resistance mechanisms, we assessed ctDNA at baseline, at week 7, and at progression. Our next-generation sequencing assay covered all NOTCH1 mutations identified in patients’ tissues. Previously identified NOTCH1 mutations in tumor tissue were detected in ctDNA collected at baseline in 5 of 6 patients (83.3%) with a median VAF of 8.3% (range 0.3-26.9%). Two patients with detectable NOTCH1 had plasma samples collected at progression. Patient 1 had a similar VAF at baseline (14.7%) as at progression (12.4%). This patient had stable disease in target lesions (4% increase by RECIST) but developed a new brain metastasis, which likely did not result in increased shedding of ctDNA owing to the blood-brain barrier.16 Patient 2 showed an increase in VAF over time from 8.3% at baseline to 14.4% at week 7 to 56.2% at progression (24 weeks), consistent with increasing tumor burden reflected on imaging. Mutations that were detected at time of progression but not at baseline included DNMT3A P904L, PIK3CA E542K, KIT M541L, BRAF K601E, BRAF V600A, BRAF I592M, ATM A1742P, and TP53 G245D. However, there was no overlap in these novel mutations between patients 1 and 2.
Using our novel algorithm, all tumors from study patients had either truncating or missense NOTCH1 mutations predicted to be LOF.2 We measured NOTCH1 activity by quantitation of cl-NOTCH1 using IHC. Ligand binding to NOTCH1 exposes it to stepwise cleavage, releasing intracellular cl-NOTCH1 that translocates to the nucleus and alters gene expression. The anti-cl-NOTCH1 antibody12 recognizes an epitope on the intracellular NOTCH1 fragment that becomes accessible only when NOTCH1 is cleaved and does not recognize the NOTCH1 pro-form or inactive heterodimers prior to ligand activation and receptor cleavage.
We examined whether the staining patterns for cl-NOTCH1 in patient tumor biopsies were consistent with LOF. We used the wt NOTCH1-knockout FADU HNSCC cell line with doxycycline-inducible cl-NOTCH1 expression as both a negative and positive control. As expected, cl-NOTCH1 staining was not detectable in knockout FADU in the absence of doxycycline, but strong nuclear staining was induced by doxycycline. In agreement with previous work,13 nuclear staining for cl-NOTCH1 was observed sporadically throughout the basal and immediate suprabasal layer in nonmalignant adjacent tonsil tissue from patient biopsies. However, staining for cl-NOTCH1 was not apparent in tumor regions from any of the study patients examined, consistent with NOTCH1 LOF mutations.
Pharmacokinetics and Pharmacodynamics
Variable | Day 1 (n = 6) | Day 8 (n = 6) | Day 22 (n = 5) | Day 43 (n = 4) | Day 64 (n = 5) |
---|---|---|---|---|---|
C max, ng/mL | 574.5 | 574 | 691 | 525.5 | 546 |
Trough, ng/mL | 62 | 101 | 11.6 | 139 | |
C max accumulation ratio | 0.78 | 1.18 | 0.82 | 0.94 | |
Trough accumulation ratio | 1.11 | 0.94 | 0.63 |
Result of Pharmacodynamic Analysis
Seven patients consented to PK studies, although patient 9 had only the day 64 PK draw owing to SARS-CoV-2 pandemic restrictions for research blood draws. The observed median Cmax concentrations were similar across all 5 collection days: 574.5, 574, 691, 525.5, and 546 ng/mL on days 1, 8, 22, 43, and 64, respectively (P = .37). Median trough concentrations were 62, 101, 11.6, and 139 ng/mL on days 8, 22, 43, and 64, respectively, but the differences were not statistically significant (P = .66). Accumulation ratios for the Cmax and trough values were calculated for each subsequent treatment day and compared with baseline (day 1 for Cmax and day 8 for trough). Median values for the accumulation ratios were similar over time (Cmax: P = .72; trough: P = .65). We did not formally compare clinical response with PK values, but bimiralisib levels (Cmax and trough) did not appear to be any higher in patients with a response (patients 4 and 9) or prolonged stable disease (patients 1 and 2) than in those with progression (patients 5 and 6).
Assessment, Analysis, and Discussion
Completion | Study completed |
---|---|
Investigator’s Assessment | Active and should be pursued further |
Head and neck squamous cell carcinoma (HNSCC) is common and lethal despite recent advances with immunotherapy.17-20 Effective targeted therapies are still needed. A major challenge to personalized treatment for HNSCC based on genomic profiling is that most alterations are in tumor suppressors, including NOTCH1, which is mutated in ~20% of patients.2,21-24 The pattern of NOTCH1 mutations in HNSCC is consistent with LOF and supports the role of NOTCH1 as a tumor suppressor.2 There are no molecularly-targeted therapies approved for NOTCH1 mutant HNSCC, representing a significant unmet clinical need.
The PI3K/AKT/mTOR signaling pathway is one of the most frequently dysregulated pathways in cancer, including HNSCC.15,25,26 Only a few drugs that target the PI3K/AKT/mTOR pathway have been approved for clinical use15 and none for HNSCC. One reason for this lack of progress is a lack of any biomarker that predicts PI3K/AKT/mTOR pathway inhibitor activity, with the exception of the PIK3CA mutation in breast cancer.27 In other tumor types, PI3K/AKT/mTOR inhibitors have had limited clinical success even in the context of PIK3CA mutations.15,28
To address this gap, we have shown that HNSCC cell lines with NOTCH1 LOF mutations exhibit greater sensitivity to PI3K/mTOR inhibitors than their wild-type counterparts.29 PI3K/mTOR inhibition induced apoptosis in HNSCC cell lines harboring NOTCH1 LOF mutations. In contrast, HNSCC cell lines with PIK3CA mutations underwent cell cycle arrest,30 but not apoptosis, when treated with PI3K/mTOR inhibitors. In addition, PI3K/mTOR inhibitors reduced tumor growth in xenograft NOTCH1-mutant HNSCC models. In a phase I basket study of single-agent bimiralisib, one patient with NOTCH1-mutant HNSCC was treated, and this heavily pretreated patient experienced a partial response in thoracic metastases lasting for 9 months.31
Bimiralisib is a pan-class I PI3K/mTOR antagonist that potently inhibits PI3Kα and mTOR, with less potency against PI3Kβ. Bimiralisib does not significantly inhibit other protein kinases in biochemical assays32 and has a toxicity profile similar to other PI3K inhibitors.8,11 Pharmacodynamic data showed marked decreases of pathway targets in tumor tissue at therapeutic doses.11 Clinical responses and stable disease have been observed with bimiralisib.10,11
In patients with R/M HNSCC who had disease progression during cisplatin-based chemotherapy and immunotherapy, bimiralisib had an objective response rate of 17%, with a median OS and PFS of 7 and 5 months respectively. Although this trial was small, outcomes were better than the standard of care, which has an objective response rate of 5.8%, a median OS of 5.1 months, and PFS of 2.7 months (ie, the standard-of-care arm of reference19). However, the standard-of-care values do fall well within our 95% CIs.
The most significant limitation of the current study was that the SARS-CoV-2 pandemic adversely affected accrual, limited research blood collections, and led to sponsor insolvency and early termination. Additionally, patients with R/M HNSCC generally have high morbidity and mortality, as was evident not only in our bimiralisib-treated patients but also by two deaths in patients with a performance status < 2 who died from disease progression after consent but prior to study treatment. Collectively, these limitations led to a small number of evaluable patients.
The bimiralisib PK values in the current study are consistent with other published studies, which showed that the Cmax value following administration of a single 120-mg dose was 876 ± 694 ng/mL.8-11,31 Given that the Cmax concentrations proportionally increased with increasing doses up to 140 mg,31 we can compare PK data using the 150-mg dose in previous studies with PK data using the 140-mg dose in the current study. Wicki et al reported that the highest Cmax was 998 ng/mL following a dose of 150 mg,11 which is similar to our maximum Cmax value of 970 ng/mL, and the median Cmax value did not differ significantly among treatment days tested. These Cmax values are well above levels needs to inhibit PI3K and kill NOTCH1 mutant HNSCC cells.29 In our study, the intermittent dosing regimen allowed for partial clearance of the drug, and thus Cmax accumulation was not observed. With a terminal half-life of 40 h, there were detectable trough concentrations throughout the treatment cycle in all patients; these trough concentrations reached a maximal value on day 64 but did not change significantly from day 8. A caveat of our PK data is that the Cmax levels in our cohort were determined using a single time point at 1 hr after the oral dose and thus may not be reflective of the true Cmax concentrations. Although the maximum plasma concentration in most humans occurs at 1-2 h after a dose, it can occur as late as 24 h after oral administration.11 However, our study design minimized PK variability driven by variable absorption depending on gastric pH and variable elimination via CYP1A2; concomitant drugs that affect gastric pH and CYP1A2 were not allowed.
Molecular testing of plasma-derived ctDNA can be used to detect the underlying tumor molecular profile and monitor its dynamic changes over time.33 In our study, we were able to detect NOTCH1 mutations in ctDNA collected at baseline in 83% of patients, which is consistent with previously published data for targeted next-generation sequencing.34 Dynamic changes in the ctDNA quantity during treatment were consistent with the clinical course. In addition, ctDNA samples collected at progression showed new emerging molecular alterations in major cancer genes such as PIK3CA, BRAF, TP53, and others; however, a small sample size precludes any definite conclusions.
The current study is the first clinical trial to test the therapeutic vulnerability of NOTCH1-mutant HNSCC to any class of drugs. Although the trial was small, the responses and prolonged stable disease are promising in a group of HNSCC patients with a very poor prognosis. These results, if confirmed in a larger trial, may inform the development of the first targeted therapy for NOTCH1 mutant HNSCC. Because NOTCH1 LOF mutations are common in other squamous cell carcinomas, including those of the skin, esophagus, and lung; our findings may translate beyond HNSCC.35-38
Acknowledgments
This translational research was supported by The National Institutes of Health (1R01CA235620) and Cancer Prevention and Research Institute of Texas (RP200369). Statistical analyses and circulating tumor DNA studies were supported by the National Cancer Institute through MD Anderson’s Cancer Center Core Grants (P30CA016672 and CA016672). PIQUR Therapeutics AG (Basel, Switzerland) sponsored the clinical trial. We thank Erica Goodoff in the Research Medical Library at MD Anderson for editing the manuscript.
Contributor Information
Faye M Johnson, Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA.
Filip Janku, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Mohamed A Gouda, Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Hai T Tran, Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Jitesh D Kawedia, Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Pharmacy Pharmacology Research, Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Debora Schmitz, PIQUR Therapeutics AG, Basel, Switzerland.
Hendrik Streefkerk, PIQUR Therapeutics AG, Basel, Switzerland.
J Jack Lee, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Clark R Andersen, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Defeng Deng, Department of Otolaryngology, Baylor College of Medicine, Houston, TX, USA.
Seema Rawal, Department of Otolaryngology, Baylor College of Medicine, Houston, TX, USA.
Pooja A Shah, Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Adel K El-Naggar, Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Jason M Johnson, Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Mitchell J Frederick, Department of Otolaryngology, Baylor College of Medicine, Houston, TX, USA.
Conflict of Interest
Faye M. Johnson: Trovagene, Takeda (RF); Filip Janku: Mersana Therapeutics, Flame Bio, Cardiff Oncology, MedinCell, Immunomet (C/A), Astex, Novartis, BioMed Valley Discoveries, Fore Bio, Deciphera, Bristol-Myers Squibb, Asana, Ideaya Biosciences, Sanofi, Merck, F-star, JSI Innopharm, Bioxcel, Lilly, Bicara, PureTech Health, FujiFilm Pharmaceuticals, Sotio, Synlogic, NextCure, Hutchinson Medipharma (RF), Ideaya Biosciences, Synlogic, Sotio, Puretech Health, Deciphera, Crown Bioscience, Asana, Fore Bio, Novartis, Bicara, PegaOne (SAB), Cardiff Oncology, Monte Rosa Therapeutics (OI); Debora Schmitz: PIQUR Therapeutics (E, at the time of study conduct); Hendrik Streefkerk: PIQUR Therapeutics (E, at the time of study conduct), CellProthera S.A.S. (Mulhouse, France), Deciphera Pharmaceuticals, LLC (Waltham, MA, US), Nuvalent, Inc. (Cambridge, MA, US), Renal 2 Corp. (New York, NY, US), Renal 3 Corp. (New York, NY, US), Moonlake Immunotherapeutics AG (Zug, Switzerland), Omeicos GmbH (Berlin, Germany) (C/A). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board.
Data Availability
The data underlying this article are available in the article and in its online supplementary material.
References
- 1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: Revised recist guideline (version 1.1). Eur J Cancer. 2009;45:228-247. 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
- 2. Shah PA, Huang C, Li Q, et al. Notch1 signaling in head and neck squamous cell carcinoma. Cells 2020;9:2677. 10.3390/cells9122677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Choi Y, Chan AP.. Provean web server: A tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics 2015;31:2745-2747. 10.1093/bioinformatics/btv195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ng PC, Henikoff S.. Sift: Predicting amino acid changes that affect protein function. Nucleic Acids Res. 2003;31:3812-3814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Agresti A, Coull BA.. Approximate is better than “exact” for interval estimation of binomial proportions. Am Stat. 1998;52:119-126. [Google Scholar]
- 6. Kaplan EL, Meier P.. Nonparametric-estimation from incomplete observations. J Am Stat Assoc. 1958;53:457-481. 10.1080/01621459.1958.10501452. [DOI] [Google Scholar]
- 7. Therneau T and Grambsch P.. Modeling survival data: Extending the cox model. New York: Springer-Verlag; 2000 [Google Scholar]
- 8. Adjei AA, Dy GK, Zhao YJ, et al. A phase 1 study of the pi3k/mtor inhibitor pqr309 evaluating safety, pharmacokinetics (pk) and pharmacodynamics (pd) in patients (pts) with advanced solid tumors. J Clin Oncol. 2016;34:2560-2560.27185851 [Google Scholar]
- 9. Machacek M, Renaud L, Dimitrijevic S, et al. Population pharmacokinetics of pqr309, a dual pi3k/mtor inhibitor in adult patients with advanced solid tumors. Presented at 2016 ASCO Annual Meeting, Chicago, IL, 2016.
- 10. Tarantelli C, Gaudio E, Arribas AJ, et al. Pqr309 is a novel dual pi3k/mtor inhibitor with preclinical antitumor activity in lymphomas as a single agent and in combination therapy. Clin Cancer Res. 2018;24:120-129. 10.1158/1078-0432.CCR-17-1041. [DOI] [PubMed] [Google Scholar]
- 11. Wicki A, Brown N, Xyrafas A, et al. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual pi3k and mtorc1/2 inhibitor pqr309 in patients with advanced solid tumors (sakk 67/13). Eur J Cancer. 2018;96:6-16. [DOI] [PubMed] [Google Scholar]
- 12. Kluk MJ, Ashworth T, Wang H, et al. Gauging notch1 activation in cancer using immunohistochemistry. PLoS One. 2013;8:e67306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Rettig EM, Chung CH, Bishop JA, et al. Cleaved notch1 expression pattern in head and neck squamous cell carcinoma is associated with notch1 mutation, hpv status, and high-risk features. Cancer Prev Res (Phila) 2015;8:287-295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Ferrarotto R, Mitani Y, McGrail DJ, et al. Proteogenomic analysis of salivary adenoid cystic carcinomas defines molecular subtypes and identifies therapeutic targets. Clin Cancer Res. 2021;27:852-864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Janku F, Yap TA, Meric-Bernstam F.. Targeting the pi3k pathway in cancer: Are we making headway? Nat Rev Clin Oncol. 2018;15:273-291. [DOI] [PubMed] [Google Scholar]
- 16. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra-22224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209-249. [DOI] [PubMed] [Google Scholar]
- 18. Burtness B, Harrington KJ, Greil R, et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (keynote-048): A randomised, open-label, phase 3 study. Lancet 2019;394:1915-1928. [DOI] [PubMed] [Google Scholar]
- 19. Ferris RL, Blumenschein G Jr., Fayette J, et al. Nivolumab vs investigator’s choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of checkmate 141 with analyses by tumor pd-l1 expression. Oral Oncol. 2018;81:45-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Seiwert TY, Burtness B, Mehra R, et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (keynote-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17:956-965. [DOI] [PubMed] [Google Scholar]
- 21. Agrawal N, Frederick MJ, Pickering CR, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in notch1. Science 2011;333:1154-1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Stransky N, Egloff AM, Tward AD, et al. The mutational landscape of head and neck squamous cell carcinoma. Science 2011;333:1157-1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Cancer Genome Atlas N. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015;517:576-582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Morris LGT, Chandramohan R, West L, et al. The molecular landscape of recurrent and metastatic head and neck cancers: Insights from a precision oncology sequencing platform. JAMA Oncol 2017;3:244-255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Isaacsson Velho PH, Castro G Jr., Chung CH.. Targeting the pi3k pathway in head and neck squamous cell carcinoma. Am Soc Clin Oncol Educ Book 2015;123:128. [DOI] [PubMed] [Google Scholar]
- 26. Jung K, Kang H, Mehra R.. Targeting phosphoinositide 3-kinase (pi3k) in head and neck squamous cell carcinoma (hnscc). Cancers Head Neck 2018;3:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Andre F, Ciruelos EM, Juric D, et al. Alpelisib plus fulvestrant for pik3ca-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from solar-1. Ann Oncol. 2021;32:208-217. [DOI] [PubMed] [Google Scholar]
- 28. Cai Y, Dodhia S, Su GH.. Dysregulations in the pi3k pathway and targeted therapies for head and neck squamous cell carcinoma. Oncotarget 2017;8:22203-22217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Sambandam V, Frederick MJ, Shen L, et al. Pdk1 mediates notch1-mutated head and neck squamous carcinoma vulnerability to therapeutic pi3k/mtor inhibition. Clin Cancer Res. 2019;25:3329-3340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Mazumdar T, Byers LA, Ng PK, et al. A comprehensive evaluation of biomarkers predictive of response to pi3k inhibitors and of resistance mechanisms in head and neck squamous cell carcinoma. Mol Cancer Ther. 2014;13:2738-2750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Janku F, Johnson FM, Opyrchal M, et al. Oral dual pi3k/mtor inhibitor bimiralisib demonstrates tolerability and a signal of activity in head and neck squamous cell cancer with notch1 loss-of-function mutation. Presented at AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics, Boston, MA, 2019.
- 32. Beaufils F, Cmiljanovic N, Cmiljanovic V, et al. 5-(4,6-dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (pqr309), a potent, brain-penetrant, orally bioavailable, pan-class i pi3k/mtor inhibitor as clinical candidate in oncology. J Med Chem. 2017;60:7524-7538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Polivka J Jr., Pesta M, Janku F.. Testing for oncogenic molecular aberrations in cell-free DNA-based liquid biopsies in the clinic: Are we there yet? Expert Rev Mol Diagn. 2015;15:1631-1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Janku F, Zhang S, Waters J, et al. Development and validation of an ultradeep next-generation sequencing assay for testing of plasma cell-free DNA from patients with advanced cancer. Clin Cancer Res. 2017;23:5648-5656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Pickering CR, Zhou JH, Lee JJ, et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin Cancer Res. 2014;20:6582-6592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Agrawal N, Jiao Y, Bettegowda C, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov 2012;2:899-905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Martincorena I, Fowler JC, Wabik A, et al. Somatic mutant clones colonize the human esophagus with age. Science 2018;362:911-917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012;489:519-525. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data underlying this article are available in the article and in its online supplementary material.