High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma

A Miller, Y Asmann, L Cattaneo, E Braggio, J Keats, D Auclair, S Lonial, MMRF CoMMpass Network, S J Russell, A K Stewart, A Miller, Y Asmann, L Cattaneo, E Braggio, J Keats, D Auclair, S Lonial, MMRF CoMMpass Network, S J Russell, A K Stewart

Abstract

Tumor-specific mutations can result in immunogenic neoantigens, both of which have been correlated with responsiveness to immune checkpoint inhibitors in highly mutagenic cancers. However, early results of single-agent checkpoint inhibitors in multiple myeloma (MM) have been underwhelming. Therefore, we sought to understand the relationship between mutation and neoantigen landscape of MM patients and responsiveness to therapies. Somatic mutation burden, neoantigen load, and response to therapy were determined using interim data from the MMRF CoMMpass study (NCT01454297) on 664 MM patients. In this population, the mean somatic and missense mutation loads were 405.84(s=608.55) and 63.90(s=95.88) mutations per patient, respectively. There was a positive linear relationship between mutation and neoantigen burdens (R2=0.862). The average predicted neoantigen load was 23.52(s=52.14) neoantigens with an average of 9.40(s=26.97) expressed neoantigens. Survival analysis revealed significantly shorter progression-free survival (PFS) in patients with greater than average somatic missense mutation load (N=163, 0.493 vs 0.726 2-year PFS, P=0.0023) and predicted expressed neoantigen load (N=214, 0.555 vs 0.729 2-year PFS, P=0.0028). This pattern is maintained when stratified by disease stage and cytogenetic abnormalities. Therefore, high mutation and neoantigen load are clinically relevant risk factors that negatively impact survival of MM patients under current standards of care.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Patient-specific mutational load. (a) Distribution of individual patient missense, nonsense, and frameshift somatic mutation counts for the 664 patient cohort. Box represents 25–75% of patients. Bolded line marks the median mutation count. (b) Histogram of natural log-transformed somatic missense mutation count shows log transformation normalizes the distribution of missense mutation frequency in patients.
Figure 2
Figure 2
Correlation between somatic missense mutation load and predicted neoantigen load. (a) Example of predicted neoantigen-binding affinities relative to wild-type binding affinities in a patient with near average mutation load (missense mutation load=64). Predicted binding affinity (IC50) for every 8/9/10-mer peptide containing somatic missense mutation compared to the predicted binding affinity of the consensus non-mutated peptide. (b) Predicted neoantigen-binding affinities relative to wild-type binding affinities for the same patient after limiting the x axis to 500 nM. The red line indicates the accepted cutoff for potential neoantigens. Points above the red line are those with mutant peptide-binding affinity IC50<500 nM and wild-type peptide-binding affinity IC50>500 nM, and are considered potential neoantigens. (c) Missense mutation load vs number of potential neoantigens with predicted binding affinity IC50<500 nM and consensus wild-type-binding affinity IC50>500 nM for all multiple myeloma test cohort patients (linear regression analysis R2=0.862). (d) Magnified view of (c) using decreased x- and y axis limits to show missense mutation vs predicted neoantigen load in test cohort. (e) Histogram of natural log-transformed predicted neoantigen count (mutant peptide-binding affinity IC50<500 nM and wild-type peptide-binding affinity IC50>500 nM) shows log transformation normalizes the distribution of predicted neoantigen frequency in patients. (f) Histogram of natural log-transformed predicted expressed neoantigen count (mutant peptide-binding affinity IC50<500 nM, wild-type peptide-binding affinity IC50>500 nM, RNAseq counts ⩾1) shows log transformation normalizes the distribution of predicted expressed neoantigen frequency in patients.
Figure 3
Figure 3
Survival in multiple myeloma patients stratified by mutation and neoantigen load. Kaplan–Meier survival curves for 664 multiple myeloma test cohort patients showing (a) length of overall survival (days) and (b) length of progression-free survival (days) after induction therapy. Univariate Kaplan–Meier survival comparison of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients with (c) below average (low) or above average (high) somatic mutation load, (d) below average (low) or above average (high) neoantigens with predicted binding affinity IC50<500 nM and consensus wild-type binding affinity IC50>500 nM, (e) below average (low) or above average (high) expressed (RNAseq counts ≥1) neoantigens with predicted binding affinity IC50<500 nM and consensus wild-type binding affinity IC50>500 nM, and (f) both high somatic mutation load and high expressed neoantigen load vs those with either low somatic mutation load, low expressed neoantigen load, or both. (g)Cox-proportional hazard rate and corresponding forest plot associated with progression-free survival of multiple myeloma test cohort patients based on mutation burden and neoantigen burden thresholds as Cox analysis variables.
Figure 4
Figure 4
Survival in bortezomib-treated and IMID-treated multiple myeloma patients stratified by mutation and neoantigen load. Kaplan–Meier survival curves for patients from test cohort receiving bortezomib as part of first-line therapy showing length of progression-free survival (days) after induction therapy in patients with (a) below average (low) or above average (high) somatic mutation load, (b) below average (low) or above average (high) expressed (RNAseq counts ⩾1) neoantigens with predicted binding affinity IC50<500 nM and consensus wild-type-binding affinity IC50>500 nM. Kaplan–Meier survival curves for patients from test cohort receiving IMIDs (lanalidomide, pomalidomide, thalidomide) as part of first-line therapy showing length of progression-free survival (days) after induction therapy in patients with (a) below average (low) or above average (high) somatic mutation load, (b) below average (low) or above average (high) expressed (RNAseq counts ⩾1) neoantigens with predicted binding affinity IC50<500 nM and consensus wild-type-binding affinity IC50>500 nM.
Figure 5
Figure 5
Relationship between somatic missense mutation burden, predicted neoantigen load, ISS stage and survival. (a) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients separated by ISS disease stage. Boxplots showing distribution of (b) missense mutation load and (c) predicted expressed neoantigen load in patients with ISS stage I, stage II, or stage III disease. (d) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients separated by ISS disease stage and below average (low) or above average (high) somatic missense mutation burden. (e) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients separated by ISS disease stage and below average (low) or above average (high) expressed neoantigen load.
Figure 6
Figure 6
Cox-proportional hazard rate and corresponding forest plot associated with progression-free survival of multiple myeloma test cohort patients based on univariate and multivariate comparisons of disease stage, cytogenetic abnormality, and mutation and neoantigen burden as Cox analysis variables.
Figure 7
Figure 7
Relationship between somatic missense mutation burden, predicted neoantigen load, cytogenetic MM subtype, and survival. (a) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients with high-risk (t(4:14), t(14:16), t(14:20) translocations, or deletion 17p) or low-risk cytogenetic abnormalities. Boxplots showing distribution of (b) missense mutation load (P=0.005) and (c) predicted expressed neoantigen load in patients with high-risk or low-risk cytogenetic abnormalities (P=0.018). (d) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients with low-risk or high-risk cytogenetic abnormalities and below average (low) or above average (high) somatic missense mutation burden. (e) Kaplan–Meier survival curves showing length of progression-free survival (days) after induction therapy in multiple myeloma test cohort patients low-risk or high-risk cytogenetic abnormalities with below average (low) or above average (high) expressed neoantigen load.

References

    1. Braggio E, Kortum KM, Stewart AK. SnapShot: Multiple Myeloma. Cancer Cell 2015; 28: e671.
    1. Rajkumar SV. Multiple myeloma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol 2016; 91: 719–734.
    1. Russell SJ, Rajkumar SV. Multiple myeloma and the road to personalised medicine. Lancet Oncol 2011; 12: 617–619.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008; 26: 677–704.
    1. Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer-preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol 2010; 37: 430–439.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–264.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015; 348: 69–74.
    1. Nielsen M, Lundegaard C, Worning P, Lauemoller SL, Lamberth K, Buus S et al. Reliable prediction of T-cell epitopes using neural networks with novel sequence representations. Protein Sci 2003; 12: 1007–1017.
    1. Andreatta M, Nielsen M. Gapped sequence alignment using artificial neural networks: application to the MHC class I system. Bioinformatics 2016; 32: 511–517.
    1. Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013; 499: 214–218.
    1. Walker BA, Wardell CP, Murison A, Boyle EM, Begum DB, Dahir NM et al. APOBEC family mutational signatures are associated with poor prognosis translocations in multiple myeloma. Nat Commun 2015; 6: 6997.
    1. Walker BA, Boyle EM, Wardell CP, Murison A, Begum DB, Dahir NM et al. Mutational spectrum, copy number changes, and outcome: results of a sequencing study of patients with newly diagnosed myeloma. J Clin Oncol 2015; 33: 3911–3920.
    1. Haricharan S, Bainbridge MN, Scheet P, Brown PH. Somatic mutation load of estrogen receptor-positive breast tumors predicts overall survival: an analysis of genome sequence data. Breast Cancer Res Treat 2014; 146: 211–220.
    1. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128.
    1. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015; 350: 207–211.
    1. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E et al. Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood 2014; 124: 453–462.
    1. Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M et al. Preliminary Results of a Phase I Study of Nivolumab (BMS-936558) in Patients with Relapsed or Refractory Lymphoid Malignancies. Blood 2014; 124: 291–291.
    1. Mateos M-V, Orlowski RZ, Siegel DSD, Reece DE, Moreau P, Ocio EM et al. Pembrolizumab in combination with lenalidomide and low-dose dexamethasone for relapsed/refractory multiple myeloma (RRMM): Final efficacy and safety analysis. J Clin Oncol 2016; 34: 8010.
    1. Badros AZ, Hyjek E, Ma N, Lesokhin AM, Rapoport AP, Kocoglu MH et al. pembrolizumab in combination with pomalidomide and dexamethasone for relapsed/refractory multiple myeloma (RRMM). Am Soc Hematol 2016, Abstract 490.
    1. Craig DW, Liang W, Venkata Y, Kurdoglu A, Aldrich J, Auclair D et al. Interim analysis of the mmrf commpass trial, a longitudinal study in multiple myeloma relating clinical outcomes to genomic and immunophenotypic profiles. Blood 2013; 122: 532–532.
    1. Lonial S, Yellapantula VD, Liang W, Kurdoglu A, Aldrich J, Legendre CM et al. Interim Analysis of the Mmrf Commpass Trial: identification of novel rearrangements potentially associated with disease initiation and progression. Blood 2014; 124: 722–722.
    1. Keats JJ, Speyer G, Christofferson A, Legendre C, Aldrich J, Russell M et al. Molecular predictors of outcome and drug response in multiple myeloma: An Interim Analysis of the Mmrf CoMMpass Study. Blood 2016; 128: 194–194.
    1. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. 2013, arXiv:1303.3997 [q-bio].
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N et al. The sequence alignment/map format and SAMtools. Bioinformatics 2009; 25: 2078–2079.
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010; 20: 1297–1303.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009; 25: 1754–1760.
    1. Saunders CT, Wong WS, Swamy S, Becq J, Murray LJ, Cheetham RK. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 2012; 28: 1811–1817.
    1. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013; 31: 213–219.
    1. Christoforides A, Carpten JD, Weiss GJ, Demeure MJ, Von Hoff DD, Craig DW. Identification of somatic mutations in cancer through Bayesian-based analysis of sequenced genome pairs. BMC Genomics 2013; 14: 302.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29: 15–21.
    1. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 2012; 7: 562–578.
    1. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 2015; 31: 166–169.
    1. Shukla SA, Rooney MS, Rajasagi M, Tiao G, Dixon PM, Lawrence MS et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol 2015; 33: 1152–1158.
    1. Therneau T2015A Package for Survival Analysis in R. version 2.38 .
    1. Therneau TM, Grambsch PM. Modeling Survival Data: Extending the Cox Model. Springer: New York, NY, USA, 2000.
    1. R Core Team. R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing: Vienna, Austria, 2015.
    1. Sidney J, Southwood S, Oseroff C, del Guercio MF, Sette A, Grey HM. Measurement of MHC/peptide interactions by gel filtration. Curr Protoc Immunol. John Wiley & Sons, Ltd, 2001; Chapter 18: a006957.
    1. Xing Y, Hogquist KA. T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol 2012; 4 doi:10.1101/cshperspect.a006957.
    1. Garcia-Munoz R, Llorente L. Chronic lymphocytic leukaemia: could immunological tolerance mechanisms be the origin of lymphoid neoplasms? Immunology 2014; 142: 536–550.
    1. Greipp PR, San Miguel J, Durie BG, Crowley JJ, Barlogie B, Blade J et al. International staging system for multiple myeloma. J Clin Oncol 2005; 23: 3412–3420.
    1. Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK et al. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia 2009; 23: 2210–2221.
    1. Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple myeloma for clinical practice. Blood Cancer J 2015; 5: e365.
    1. Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M et al. Prediction and prioritization of neoantigens: integration of RNA-Seq data with whole-exome sequencing. Cancer Sci 2017; 108: 170–177.
    1. Hundal J, Carreno BM, Petti AA, Linette GP, Griffith OL, Mardis ER et al. pVAC-Seq: A genome-guided in silico approach to identifying tumor neoantigens. Genome Med 2016; 8: 11.
    1. Vita R, Overton JA, Greenbaum JA, Ponomarenko J, Clark JD, Cantrell JR et al. The immune epitope database (IEDB) 3.0. Nucleic Acids Res 2015; 43: D405–D412.
    1. Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L et al. Revised international staging system for multiple myeloma: A Report From International Myeloma Working Group. J Clin Oncol 2015; 33: 2863–2869.
    1. Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, Smyth MJ et al. Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia 2010; 24: 22–32.
    1. Shi J, Tricot GJ, Garg TK, Malaviarachchi PA, Szmania SM, Kellum RE et al. Bortezomib down-regulates the cell-surface expression of HLA class I and enhances natural killer cell-mediated lysis of myeloma. Blood 2008; 111: 1309–1317.
    1. Walker BA, Wardell CP, Melchor L, Brioli A, Johnson DC, Kaiser MF et al. Intraclonal heterogeneity is a critical early event in the development of myeloma and precedes the development of clinical symptoms. Leukemia 2014; 28: 384–390.
    1. Chapman MA, Lawrence MS, Keats JJ, Cibulskis K, Sougnez C, Schinzel AC et al. Initial genome sequencing and analysis of multiple myeloma. Nature 2011; 471: 467–472.
    1. Bolli N, Avet-Loiseau H, Wedge DC, Van Loo P, Alexandrov LB, Martincorena I et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun 2014; 5: 2997.

Source: PubMed

3
Předplatit