The PEMDAC phase 2 study of pembrolizumab and entinostat in patients with metastatic uveal melanoma

Lars Ny, Henrik Jespersen, Joakim Karlsson, Samuel Alsén, Stefan Filges, Charlotta All-Eriksson, Bengt Andersson, Ana Carneiro, Hildur Helgadottir, Max Levin, Ingrid Ljuslinder, Roger Olofsson Bagge, Vasu R Sah, Ulrika Stierner, Anders Ståhlberg, Gustav Ullenhag, Lisa M Nilsson, Jonas A Nilsson, Lars Ny, Henrik Jespersen, Joakim Karlsson, Samuel Alsén, Stefan Filges, Charlotta All-Eriksson, Bengt Andersson, Ana Carneiro, Hildur Helgadottir, Max Levin, Ingrid Ljuslinder, Roger Olofsson Bagge, Vasu R Sah, Ulrika Stierner, Anders Ståhlberg, Gustav Ullenhag, Lisa M Nilsson, Jonas A Nilsson

Abstract

Preclinical studies have suggested that epigenetic therapy could enhance immunogenicity of cancer cells. We report the results of the PEMDAC phase 2 clinical trial (n = 29; NCT02697630) where the HDAC inhibitor entinostat was combined with the PD-1 inhibitor pembrolizumab in patients with metastatic uveal melanoma (UM). The primary endpoint was objective response rate (ORR), and was met with an ORR of 14%. The clinical benefit rate at 18 weeks was 28%, median progression free survival was 2.1 months and the median overall survival was 13.4 months. Toxicities were manageable, and there were no treatment-related deaths. Objective responses and/or prolonged survival were seen in patients with BAP1 wildtype tumors, and in one patient with an iris melanoma that exhibited a UV signature. Longer survival also correlated with low baseline ctDNA levels or LDH. In conclusion, HDAC inhibition and anti-PD1 immunotherapy results in durable responses in a subset of patients with metastatic UM.Trial registration ClinicalTrials.gov registration number: NCT02697630 (registered 3 March 2016). EudraCT registration number: 2016-002114-50.

Conflict of interest statement

Lars Ny received a research grant from MSD and Syndax Pharmaceuticals to support some aspects of the PEMDAC study. Anders Ståhlberg is the coinventor of SiMSen-Seq (ctDNA measurement), is a board member of, and has stock ownership in SiMSen Diagnostics. The remaining authors declare no conflict of interest.

© 2021. The Author(s).

Figures

Fig. 1. Clinical outcome data from the…
Fig. 1. Clinical outcome data from the PEMDAC trial at data cutoff in December 2019.
a Waterfall plot showing maximum change in sum of target lesion diameter from baseline to data cutoff. One patient did not have a response assessment after baseline and is not included in the figure. Dotted lines represent thresholds for progressive disease (+20%) and partial response (−30%), respectively, according to RECIST v1.1 criteria. b Spider plot showing change in the sum of target lesion diameter over time for all patients with at least one follow-up scan. c Swimmer’s plot showing time on treatment, time to best response, and duration of response in all patients who received at least one dose of study drug. In panels (a,b), n = 28 and in (c) n = 29 patients are shown, respectively. d Kaplan–Meier analysis showing overall survival (OS) of all patients. e Kaplan–Meier analysis showing progression-free survival (PFS) of all patients except one. f Circulating tumor DNA levels in patients with UM. Plasma from twenty-five patients was analyzed for the presence of mutated reads in either GNA11 (Q209L) or GNAQ (Q209L/P). Variant allele frequencies (VAF) in percent compared with reads with wildtype alleles are plotted. g Kaplan–Meier analysis comparing OS between patients with high or low (relative to median) levels of total detected ctDNA copies, n = 14 and n = 15 patients, respectively. h Kaplan–Meier analysis comparing OS between patients with lactate dehydrogenase (LDH) baseline greater or lower than the upper limit of normal (ULN), n = 17 and n = 12 patients, respectively. In (g, h), p-values for survival associations were calculated using log-rank tests. No adjustments for multiple comparisons were made. All statistical tests were two-sided. Source data are provided as a Source Data file.
Fig. 2. Genetic analyses of pretreatment biopsies…
Fig. 2. Genetic analyses of pretreatment biopsies from patients recruited to the PEMDAC trial.
a Mutations in genes that are either recurrently altered in UM or listed among COSMIC Cancer Gene Census driver genes. Responses in the trial are indicated. b Copy number profiles of each tumor inferred from exome sequencing data of tumors and matched normal tissue. Differences in color intensity depend on copy number amplitude and tumor purity. c, d OS and PFS analyses comparing patients with GNAQ- or GNA11-mutated UM (n = 21). e Fisher’s exact test of BAP1 mutational status versus responses in the PEMDAC trial. PD: progressive disease; SD: stable disease; PR: partial response. f Number of detected mutations from exome-sequencing data. g CT scans of patient 4–022 at baseline and at best response (22 months post therapy). Arrows show PET-positive lesions that disappeared from the dorsal neck region and left gluteus. h, i Comparison of OS and PFS to assess if UM patients with a wild-type BAP1 status or a UV-damaged genome survive longer than the other patients (n = 24). In (c, d) and (h, i), p-values for survival associations were calculated using log-rank tests. No adjustments for multiple comparisons were made. All statistical tests were two-sided. Source data are provided as a Source Data file.
Fig. 3. Immune profiles of pre- and…
Fig. 3. Immune profiles of pre- and post-treatment (one cycle) blood samples.
ac Flow cytometry analyses of changes in circulating cell populations in shorter and longer survivors following treatment with entinostat and pembrolizumab. Gating strategies can be found in Supplementary Fig. 5. (a) Graphs showing the frequency of CD3 + lymphocytes (left), CD14 + monocytes (middle), and CD33 + neutrophils (right) among patients with shorter (n = 12) and (b) longer (n = 12) survival prior to (circles) or after (squares) one cycle of treatment. (c) Analysis of CD8 + cytotoxic T cells in patients with shorter and longer survival. Statistical analysis was performed using multiple paired t-tests correct for multiple comparisons using a two-stage step-up (Benjamini, Krieger, and Yekutieli) with test between all groups in (ab), where * indicates adjusted p-values < 0.05. d White blood cells from three patients were analyzed by 10x Genomics TCR and gene expression analysis. Statistically altered genes in different cell types after treatment in the categories antigen presentation, immune-checkpoint receptors, and transcriptional regulation. n = 3 patient samples were compared pre- and post treatment. Genes shown were significant at q < 0.05. p-values were calculated with Wilcoxon rank-sum tests and adjusted for multiple comparisons with the Benjamini–Hochberg method. All statistical tests were two-sided. Source data are provided as a Source Data file.

References

    1. Bergman L, et al. Incidence of uveal melanoma in Sweden from 1960 to 1998. Invest. Ophthalmol. Vis. Sci. 2002;43:2579–2583.
    1. Damato B. Treatment of primary intraocular melanoma. Expert Rev. Anticancer Ther. 2006;6:493–506. doi: 10.1586/14737140.6.4.493.
    1. Jager MJ, et al. Uveal melanoma. Nat. Rev. Dis. Prim. 2020;6:24. doi: 10.1038/s41572-020-0158-0.
    1. Kujala E, Makitie T, Kivela T. Very long-term prognosis of patients with malignant uveal melanoma. Invest. Ophthalmol. Vis. Sci. 2003;44:4651–4659. doi: 10.1167/iovs.03-0538.
    1. Diener-West M, et al. Development of metastatic disease after enrollment in the COMS trials for treatment of choroidal melanoma: Collaborative Ocular Melanoma Study Group Report No. 26. Arch. Ophthalmol. 2005;123:1639–1643. doi: 10.1001/archopht.123.12.1639.
    1. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082.
    1. Robert C, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015;372:2521–2532. doi: 10.1056/NEJMoa1503093.
    1. Larkin J, et al. Five-year survival with combined Nivolumab and Ipilimumab in advanced melanoma. N. Engl. J. Med. 2019;381:1535–1546. doi: 10.1056/NEJMoa1910836.
    1. Khoja L, et al. Meta-analysis in metastatic uveal melanoma to determine progression-free and overall survival benchmarks: an international rare cancers initiative (IRCI) ocular melanoma study. Ann. Oncol. 2019;30:1370–1380. doi: 10.1093/annonc/mdz176.
    1. Van Raamsdonk CD, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602. doi: 10.1038/nature07586.
    1. Van Raamsdonk CD, et al. Mutations in GNA11 in uveal melanoma. N. Engl. J. Med. 2010;363:2191–2199. doi: 10.1056/NEJMoa1000584.
    1. Robertson AG, et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell. 2017;32:204–220. doi: 10.1016/j.ccell.2017.07.003.
    1. Karlsson J, et al. Molecular profiling of driver events in metastatic uveal melanoma. Nat. Commun. 2020;11:1894. doi: 10.1038/s41467-020-15606-0.
    1. Chandran SS, et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017;18:792–802. doi: 10.1016/S1470-2045(17)30251-6.
    1. Middleton MR, et al. Tebentafusp, a TCR/anti-CD3 bispecific fusion protein targeting gp100, potently activated anti-tumor immune responses in patients with metastatic melanoma. Clin. Cancer Res. 2020;26:5869–5878. doi: 10.1158/1078-0432.CCR-20-1247.
    1. Algazi AP, et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-1L antibodies. Cancer. 2016;122:3344–3353. doi: 10.1002/cncr.30258.
    1. Mignard C, et al. Efficacy of immunotherapy in patients with metastatic mucosal or uveal melanoma. J. Oncol. 2018;2018:1908065. doi: 10.1155/2018/1908065.
    1. Pelster MS, et al. Nivolumab and Ipilimumab in metastatic uveal melanoma: results from a single-arm phase II study. J. Clin. Oncol. 2021;39:599–607. doi: 10.1200/JCO.20.00605.
    1. Piulats JM, et al. Nivolumab Plus Ipilimumab for treatment-naive metastatic uveal melanoma: an open-label, multicenter, phase II trial by the Spanish Multidisciplinary Melanoma Group (GEM-1402) J. Clin. Oncol. 2021;39:586–598. doi: 10.1200/JCO.20.00550.
    1. Royer-Bertrand B, et al. Comprehensive genetic landscape of uveal melanoma by whole-genome sequencing. Am. J. Hum. Genet. 2016;99:1190–1198. doi: 10.1016/j.ajhg.2016.09.008.
    1. Whelchel JC, Farah SE, McLean IW, Burnier MN. Immunohistochemistry of infiltrating lymphocytes in uveal malignant melanoma. Invest. Ophthalmol. Vis. Sci. 1993;34:2603–2606.
    1. Maat W, et al. Monosomy of chromosome 3 and an inflammatory phenotype occur together in uveal melanoma. Invest. Ophthalmol. Vis. Sci. 2008;49:505–510. doi: 10.1167/iovs.07-0786.
    1. Makitie T, Summanen P, Tarkkanen A, Kivela T. Tumor-infiltrating macrophages (CD68(+) cells) and prognosis in malignant uveal melanoma. Invest. Ophthalmol. Vis. Sci. 2001;42:1414–1421.
    1. Topper MJ, Vaz M, Marrone KA, Brahmer JR, Baylin SB. The emerging role of epigenetic therapeutics in immuno-oncology. Nat. Rev. Clin. Oncol. 2020;17:75–90. doi: 10.1038/s41571-019-0266-5.
    1. Shen L, Pili R. Class I histone deacetylase inhibition is a novel mechanism to target regulatory T cells in immunotherapy. Oncoimmunology. 2012;1:948–950. doi: 10.4161/onci.20306.
    1. Kim K, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc. Natl Acad. Sci. USA. 2014;111:11774–11779. doi: 10.1073/pnas.1410626111.
    1. Maio M, Coral S, Fratta E, Altomonte M, Sigalotti L. Epigenetic targets for immune intervention in human malignancies. Oncogene. 2003;22:6484–6488. doi: 10.1038/sj.onc.1206956.
    1. Landreville S, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clin. Cancer Res. 2012;18:408–416. doi: 10.1158/1078-0432.CCR-11-0946.
    1. Lee JH, Choy ML, Ngo L, Foster SS, Marks PA. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc. Natl Acad. Sci. USA. 2010;107:14639–14644. doi: 10.1073/pnas.1008522107.
    1. Campoli M, Ferrone S. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 2008;27:5869–5885. doi: 10.1038/onc.2008.273.
    1. Sah, V. R. et al. Epigenetic therapy to enhance therapeutic effects of PD-1 inhibition in uveal melanoma. bioRxiv, 10.1101/2021.02.04.429575 (2021).
    1. Woods DM, et al. HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol. Res. 2015;3:1375–1385. doi: 10.1158/2326-6066.CIR-15-0077-T.
    1. Gao Y, et al. Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat. Cell Biol. 2020;22:1064–1075. doi: 10.1038/s41556-020-0562-4.
    1. Christmas BJ, et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol. Res. 2018;6:1561–1577. doi: 10.1158/2326-6066.CIR-18-0070.
    1. Orillion A, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin. Cancer Res. 2017;23:5187–5201. doi: 10.1158/1078-0432.CCR-17-0741.
    1. Sullivan, R. J. et al. Abstract CT072: Efficacy and safety of entinostat (ENT) and pembrolizumab (PEMBRO) in patients with melanoma previously treated with anti-PD1 therapy. Cancer Res.79 (13 Supplement), CT072 (2019).
    1. Gandhi L, et al. Efficacy and safety of entinostat (ENT) and pembrolizumab (PEMBRO) in patients with non-small cell lung cancer (NSCLC) previously treated with anti-PD-(L)1 therapy. J. Clin. Oncol. 2018;36:9036–9036. doi: 10.1200/JCO.2018.36.15_suppl.9036.
    1. Zheng H, et al. HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma. Clin. Cancer Res. 2016;22:4119–4132. doi: 10.1158/1078-0432.CCR-15-2584.
    1. Kim YD, et al. HDAC inhibitor, CG-745, enhances the anti-cancer effect of Anti-PD-1 immune checkpoint inhibitor by modulation of the immune microenvironment. J. Cancer. 2020;11:4059–4072. doi: 10.7150/jca.44622.
    1. Gray JE, et al. Phase I/Ib study of pembrolizumab plus vorinostat in advanced/metastatic non-small cell lung cancer. Clin. Cancer Res. 2019;25:6623–6632. doi: 10.1158/1078-0432.CCR-19-1305.
    1. Rodriguez CP, et al. A phase II trial of pembrolizumab and vorinostat in recurrent metastatic head and neck squamous cell carcinomas and salivary gland cancer. Clin. Cancer Res. 2020;26:837–845. doi: 10.1158/1078-0432.CCR-19-2214.
    1. Johansson PA, et al. Whole genome landscapes of uveal melanoma show an ultraviolet radiation signature in iris tumours. Nat. Commun. 2020;11:2408. doi: 10.1038/s41467-020-16276-8.
    1. Bhadury J, et al. BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc. Natl Acad. Sci. USA. 2014;111:E2721–E2730. doi: 10.1073/pnas.1406722111.
    1. Eigentler TK, et al. Diagnosis, monitoring and management of immune-related adverse drug reactions of anti-PD-1 antibody therapy. Cancer Treat. Rev. 2016;45:7–18. doi: 10.1016/j.ctrv.2016.02.003.
    1. Weber JS, et al. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J. Clin. Oncol. 2017;35:785–792. doi: 10.1200/JCO.2015.66.1389.
    1. Haratani K, et al. Association of immune-related adverse events with nivolumab efficacy in non-small-cell lung cancer. JAMA Oncol. 2018;4:374–378. doi: 10.1001/jamaoncol.2017.2925.
    1. Piulats Rodriguez, J. M. et al. Phase II multicenter, single arm, open label study of Nivolumab in combination with Ipilimumab in untreated patients with metastatic uveal melanoma. Annals of Oncology. 29, viii442–viii466 (2018).
    1. Najjar YG, et al. Ipilimumab plus nivolumab for patients with metastatic uveal melanoma: a multicenter, retrospective study. J. Immunother. Cancer. 2020;8:e000331. doi: 10.1136/jitc-2019-000331.
    1. Sznol M, et al. Pooled analysis safety profile of nivolumab and ipilimumab combination therapy in patients with advanced melanoma. J. Clin. Oncol. 2017;35:3815–3822. doi: 10.1200/JCO.2016.72.1167.
    1. Nabet BY, et al. Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell. 2020;183:363–376. doi: 10.1016/j.cell.2020.09.001.
    1. Marsavela G, et al. Circulating tumor DNA predicts outcome from first-, but not second-line treatment and identifies melanoma patients who may benefit from combination immunotherapy. Clin. Cancer Res. 2020;26:5926–5933. doi: 10.1158/1078-0432.CCR-20-2251.
    1. Johnson DB, et al. Response to anti-PD-1 in uveal melanoma without high-volume liver metastasis. J. Natl Compr. Canc. Netw. 2019;17:114–117. doi: 10.6004/jnccn.2018.7070.
    1. Muller L, Mitsuhashi M, Simms P, Gooding WE, Whiteside TL. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 2016;6:20254. doi: 10.1038/srep20254.
    1. Johansson PA, et al. Prolonged stable disease in a uveal melanoma patient with germline MBD4 nonsense mutation treated with pembrolizumab and ipilimumab. Immunogenetics. 2019;71:433–436. doi: 10.1007/s00251-019-01108-x.
    1. Rodrigues M, et al. Outlier response to anti-PD1 in uveal melanoma reveals germline MBD4 mutations in hypermutated tumors. Nat. Commun. 2018;9:1866. doi: 10.1038/s41467-018-04322-5.
    1. Harbour JW, et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science. 2010;330:1410–1413. doi: 10.1126/science.1194472.
    1. Carbone M, et al. Biological mechanisms and clinical significance of BAP1 mutations in human cancer. Cancer Disco. 2020;10:1103–1120. doi: 10.1158/-19-1220.
    1. Figueiredo CR, et al. Loss of BAP1 expression is associated with an immunosuppressive microenvironment in uveal melanoma, with implications for immunotherapy development. J. Pathol. 2020;250:420–439. doi: 10.1002/path.5384.
    1. Kuznetsov JN, et al. BAP1 regulates epigenetic switch from pluripotency to differentiation in developmental lineages giving rise to BAP1-mutant cancers. Sci. Adv. 2019;5:eaax1738. doi: 10.1126/sciadv.aax1738.
    1. Sacco JJ, et al. Loss of the deubiquitylase BAP1 alters class I histone deacetylase expression and sensitivity of mesothelioma cells to HDAC inhibitors. Oncotarget. 2015;6:13757–13771. doi: 10.18632/oncotarget.3765.
    1. Matatall KA, et al. BAP1 deficiency causes loss of melanocytic cell identity in uveal melanoma. BMC Cancer. 2013;13:371. doi: 10.1186/1471-2407-13-371.
    1. Haas NB, et al. Phase II trial of vorinostat in advanced melanoma. Invest. New Drugs. 2014;32:526–534. doi: 10.1007/s10637-014-0066-9.
    1. Jespersen H, et al. Concomitant use of pembrolizumab and entinostat in adult patients with metastatic uveal melanoma (PEMDAC study): protocol for a multicenter phase II open label study. BMC Cancer. 2019;19:415. doi: 10.1186/s12885-019-5623-3.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Nishino M, et al. Developing a common language for tumor response to immunotherapy: immune-related response criteria using unidimensional measurements. Clin. Cancer Res. 2013;19:3936–3943. doi: 10.1158/1078-0432.CCR-13-0895.
    1. Simon R. Optimal two-stage designs for phase II clinical trials. Control. Clin. Trials. 1989;10:1–10. doi: 10.1016/0197-2456(89)90015-9.
    1. Stahlberg A, et al. Simple multiplexed PCR-based barcoding of DNA for ultrasensitive mutation detection by next-generation sequencing. Nat. Protoc. 2017;12:664–682. doi: 10.1038/nprot.2017.006.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. McKenna A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110.
    1. Genomes Project C, et al. A global reference for human genetic variation. Nature. 2015;526:68–74. doi: 10.1038/nature15393.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169. doi: 10.1093/bioinformatics/btu638.
    1. Benjamin, D. et al. Calling Somatic SNVs and Indels with Mutect2. bioRxiv, 10.1101/861054 (2019).
    1. Karczewski KJ, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–443. doi: 10.1038/s41586-020-2308-7.
    1. McLaren W, et al. The ensembl variant effect predictor. Genome Biol. 2016;17:122. doi: 10.1186/s13059-016-0974-4.
    1. Blokzijl F, Janssen R, van Boxtel R, Cuppen E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med. 2018;10:33. doi: 10.1186/s13073-018-0539-0.
    1. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 2016;12:e1004873. doi: 10.1371/journal.pcbi.1004873.
    1. Baran Y, et al. MetaCell: analysis of single-cell RNA-seq data using K-nn graph partitions. Genome Biol. 2019;20:206. doi: 10.1186/s13059-019-1812-2.
    1. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018;36:411–420. doi: 10.1038/nbt.4096.
    1. Lin, H. & Zelterman, D. Modeling Survival Data: Extending the Cox Model. Technometrics44, 85–86 (2002).

Source: PubMed

3
Abonner