Individualized genetic makeup that controls natural killer cell function influences the efficacy of isatuximab immunotherapy in patients with multiple myeloma

Haibo Sun, Thomas G Martin, John Marra, Denice Kong, Jonathon Keats, Sandrine Macé, Marielle Chiron, Jeffrey L Wolf, Jeffrey M Venstrom, Raja Rajalingam, Haibo Sun, Thomas G Martin, John Marra, Denice Kong, Jonathon Keats, Sandrine Macé, Marielle Chiron, Jeffrey L Wolf, Jeffrey M Venstrom, Raja Rajalingam

Abstract

Background: Phase IIb clinical trial with isatuximab (Isa)-lenalidomide (Len)-dexamethasone (Dex) showed an improved progression-free survival (PFS) in patients with relapsed or refractory multiple myeloma (RRMM), but the efficacy varied by patient. Antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) cells plays a crucial role in arbitrating antitumor activities of therapeutic-antibodies. We tested if patient-specific genetic makeup known to set NK cell functional threshold influence response to Isa-Len-Dex therapy.

Methods: We characterized 57 patients with RRMM receiving Isa-Len-Dex for polymorphisms of killer-cell immunoglobulin-like receptors (KIR), human leukocyte antigen (HLA) class I, and FCGR3A loci. In vitro ADCC assay, coincubating primary NK cells expressing specific KIR repertoire with multiple myeloma cell lines (MM cells) expressing selected HLA class I ligands, was used to confirm the identified genetic correlatives of clinical response.

Results: Patients with KIR3DL2+ and its cognate-ligand HLA-A3/11+ had superior PFS than patients missing this combination (HR=0.43; p=0.02), while patients carrying KIR2DL1+ and HLA-C2C2+ compared with to patients missing this pair showed short PFS (HR=3.54; p=0.05). Patients with KIR3DL2+ and HLA-A3/11+ plus high-affinity FCGR3A-158V allele showed the most prolonged PFS (HR=0.35; p=0.007). Consistent with these clinical data, mechanistic experiments demonstrated that NK cells expressing KIR3DL2 trigger greater ADCC when MM cells express HLA-A3/11. Inversely, NK cells expressing KIR2DL1 do not kill if MM cells express the HLA-C2C2 ligand. NK cells expressing high-affinity FCGR3A-158VV-induced greater ADCC compared with those with low-affinity FCGR3A-158FF.

Conclusions: Our results suggest that KIR3DL2+ and HLA-A3/11+ with FCGR3A-158V markers lead to enhanced Isa-dependent NK-mediated cytolysis against MM cells and results in improved PFS in patients with RRMM treated by Isa-Len-Dex. Moreover, the presence of KIR2DL1+ and HLA-C2C2+ identifies patients who may have a lower response to Isa-Len-Dex therapy linked to a reduced NK-mediated ADCC. These biomarkers could potentially identify, via precision medicine, patients more likely to respond to Isa-Len-Dex immunotherapy.

Trial registration number: NCT01749969.

Keywords: genetic markers; immunity; immunotherapy; innate.

Conflict of interest statement

Competing interests: HS: no relationship to disclose. TGM: research funding: Sanofi. JM: no relationship to disclose. DK: no relationship to disclose. JK: no relationship to disclose. SM: employee of Sanofi. MC: employee of Sanofi. JLW: consulting or advisory role: Onyx/Amgen, Takeda, and Celgene. JMV: no relationship to disclose. RR: research funding: Sanofi; consulting or advisory role: CareDx.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Killer-cell immunoglobulin-like receptors (KIR) haplotypes vary in gene content. Map of group A and B KIR haplotypes. Inhibitory KIR genes are depicted in white boxes with their specific human leukocyte antigen (HLA) class I ligands. Activating KIRs are shown in dark boxes. Hatched boxes represent pseudogenes or KIR genes with no apparent function. The centromeric and telomeric half are marked.
Figure 2
Figure 2
The Kaplan-Meier curves showing the impact of specific killer-cell immunoglobulin-like receptors (KIR)+human leukocyte antigen (HLA) combinations on progression-free survival (PFS) of relapsed or refractory multiple myeloma (RRMM) patients with RRMM treated by isatuximab+lenalidomide+dexamethasone (A, B). Patients carrying KIR3DL2+HLA-A3/11+ had significantly prolonged PFS compared with the patients missing those combinations (A). In contrast, patients with KIR2DL1+HLA-C2C2+ had significantly reduced PFS compared with patients missing this combination (B). Mechanistic studies confirm the role of these KIR+HLA interactions in modulating isatuximab-dependent natural killer (NK) cytolysis of MM cells (C, D). Multiple myeloma (MM) cells expressing HLA-A3/11 (ANBL6 expresses HLA-A3, and KMS18 expresses HLA-A11) showed a significantly improved isatuximab-dependent NK cytolysis compared with those that were negative for HLA-A3/11 (SKMM2 and JK6L) (C). All these four MM cell lines (ANBL6, KMS18, SKMM2, and JK6L) coexpress HLA-C1C1 and HLA-Bw4I80 ligands. The MM cell lines expressing HLA-C2C2 (Karpas620 and ARD) were resistant to isatuximab-dependent NK cytolysis compared with the MM cell lines (AMO1, KAS61, KMS20, and XG2) that are negative for HLA-C2C2 (D). These six MM cell lines (Karpas620, ARD, AMO1, KAS61, KMS20, and XG2) co-express HLA-Bw4T80 ligand. The cognate HLA class I ligands of the effector NK cells are given in parenthesis. Data provided in (C) and (D) represent the mean±SD of 11 independent experiments.
Figure 3
Figure 3
Impact of FCGR3A genotypes on progression-free survival (PFS) and isatuximab-dependent natural killer (NK) cell-mediated cytotoxicity. The Kaplan-Meier curves showing PFS with patients having a high-affinity FCGR3A-158VV/VF genotype compared with the patients having low-binding FCGR3A-158FF genotype (A). Six multiple myeloma (MM) cell lines expressing a similar set of human leukocyte antigen class I ligands (C1C1+Bw4+) were treated with and without isatuximab (10 µg/mL), and then exposed to primary NK cells carrying either homozygous FCGR3A-158VV or FF genotype. Effector NK cells with FCGR3A-158VV mediate greater isatuximab-dependent antibody-dependent cell-mediated cytotoxicity compared with the NK cells expressing FCGR3A-158FF (B).
Figure 4
Figure 4
The Kaplan-Meier curves showing the impact of specific killer-cell immunoglobulin like receptors (KIR)+human leukocyte antigen (HLA) and FCGR3A gene polymorphisms on progression-free survival (PFS) of patients with relapsed or refractory multiple myeloma treated by isatuximab+lenalidomide+dexamethasone. Patients carrying KIR3DL2+HLA-A3/11+ plus FCRG3A-158V (A) or KIR2DL3+HLA-C1C1+ (B) had significantly prolonged PFS compared with the patients missing those combinations.

References

    1. Röllig C, Knop S, Bornhäuser M. Multiple myeloma. Lancet 2015;385:2197–208. 10.1016/S0140-6736(14)60493-1
    1. Gozzetti A, Candi V, Papini G, et al. . Therapeutic advancements in multiple myeloma. Front Oncol 2014;4:241. 10.3389/fonc.2014.00241
    1. Michels TC, Petersen KE. Multiple myeloma: diagnosis and treatment. Am Fam Physician 2017;95:373–83.
    1. Kumar SK, Lee JH, Lahuerta JJ, et al. . Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter international myeloma Working Group study. Leukemia 2012;26:149–57. 10.1038/leu.2011.196
    1. Raje N, Longo DL. Monoclonal antibodies in multiple myeloma come of age. N Engl J Med 2015;373:1264–6. 10.1056/NEJMe1509419
    1. Lonial S, Durie B, Palumbo A, et al. . Monoclonal antibodies in the treatment of multiple myeloma: current status and future perspectives. Leukemia 2016;30:526–35. 10.1038/leu.2015.223
    1. Attal M, Richardson PG, Rajkumar SV, et al. . Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet 2019;394:2096–107. 10.1016/S0140-6736(19)32556-5
    1. Martin T, Baz R, Benson DM, et al. . A phase 1B study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood 2017;129:3294–303. 10.1182/blood-2016-09-740787
    1. van de Donk NWCJ, Richardson PG, Malavasi F. Cd38 antibodies in multiple myeloma: back to the future. Blood 2018;131:13–29. 10.1182/blood-2017-06-740944
    1. Deckert J, Wetzel M-C, Bartle LM, et al. . SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin Cancer Res 2014;20:4574–83. 10.1158/1078-0432.CCR-14-0695
    1. Jiang H, Acharya C, An G, et al. . SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 2016;30:399–408. 10.1038/leu.2015.240
    1. Moreno L, Perez C, Zabaleta A, et al. . The mechanism of action of the Anti-CD38 monoclonal antibody Isatuximab in multiple myeloma. Clin Cancer Res 2019;25:3176–87. 10.1158/1078-0432.CCR-18-1597
    1. Cerwenka A, Lanier LL. Natural killer cells, viruses and cancer. Nat Rev Immunol 2001;1:41–9. 10.1038/35095564
    1. Rajalingam R. Diversity of killer cell immunoglobulin-like receptors and disease. Clin Lab Med 2018;38:637–53. 10.1016/j.cll.2018.08.001
    1. Wilson MJ, Torkar M, Haude A, et al. . Plasticity in the organization and sequences of human KIR/ILT gene families. Proc Natl Acad Sci U S A 2000;97:4778–83. 10.1073/pnas.080588597
    1. Raulet DH, Vance RE, McMahon CW. Regulation of the natural killer cell receptor repertoire. Annu Rev Immunol 2001;19:291–330. 10.1146/annurev.immunol.19.1.291
    1. Kim S, Poursine-Laurent J, Truscott SM, et al. . Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005;436:709–13. 10.1038/nature03847
    1. Yokoyama WM, Kim S. How do natural killer cells find self to achieve tolerance? Immunity 2006;24:249–57. 10.1016/j.immuni.2006.03.006
    1. Raulet DH, Vance RE. Self-Tolerance of natural killer cells. Nat Rev Immunol 2006;6:520–31. 10.1038/nri1863
    1. Anfossi N, André P, Guia S, et al. . Human NK cell education by inhibitory receptors for MHC class I. Immunity 2006;25:331–42. 10.1016/j.immuni.2006.06.013
    1. Du Z, Gjertson DW, Reed EF, et al. . Receptor-Ligand analyses define minimal killer cell Ig-like receptor (Kir) in humans. Immunogenetics 2007;59:1–15. 10.1007/s00251-006-0168-4
    1. Khakoo SI, Carrington M. Kir and disease: a model system or system of models? Immunol Rev 2006;214:186–201. 10.1111/j.1600-065X.2006.00459.x
    1. Mellor JD, Brown MP, Irving HR, et al. . A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer. J Hematol Oncol 2013;6:1. 10.1186/1756-8722-6-1
    1. Musolino A, Naldi N, Bortesi B, et al. . Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol 2008;26:1789–96. 10.1200/JCO.2007.14.8957
    1. Taylor RJ, Chan S-L, Wood A, et al. . Fcgammariiia polymorphisms and cetuximab induced cytotoxicity in squamous cell carcinoma of the head and neck. Cancer Immunol Immunother 2009;58:997–1006. 10.1007/s00262-008-0613-3
    1. Weng W-K, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol 2003;21:3940–7. 10.1200/JCO.2003.05.013
    1. Richardson PG, Facon T, Bensinger WI, et al. . Predictive biomarkers with isatuximab plus pomalidomide and dexamethasone in relapsed/refractory multiple myeloma. Blood Cancer J 2021;11:55. 10.1038/s41408-021-00438-y
    1. Zhu C, Song Z, Wang A, et al. . Isatuximab acts through Fc-dependent, independent, and direct pathways to kill multiple myeloma cells. Front Immunol 2020;11:11. 10.3389/fimmu.2020.01771
    1. Rajalingam R, Ashouri E. Gene-Specific PCR typing of killer cell immunoglobulin-like receptors. Methods Mol Biol 2013;1034:239–55. 10.1007/978-1-62703-493-7_12
    1. Dall'Ozzo S, Andres C, Bardos P, et al. . Rapid single-step FCGR3A genotyping based on SYBR green I fluorescence in real-time multiplex allele-specific PCR. J Immunol Methods 2003;277:185–92. 10.1016/S0022-1759(03)00123-6
    1. Yamashita M, Kitano S, Aikawa H, et al. . A novel method for evaluating antibody-dependent cell-mediated cytotoxicity by flowcytometry using cryopreserved human peripheral blood mononuclear cells. Sci Rep 2016;6:19772. 10.1038/srep19772
    1. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457–81. 10.1080/01621459.1958.10501452
    1. Tallarida RJ. Quantitative methods for assessing drug synergism. Genes Cancer 2011;2:1003–8. 10.1177/1947601912440575
    1. Cella M, Longo A, Ferrara GB, et al. . NK3-specific natural killer cells are selectively inhibited by Bw4-positive HLA alleles with isoleucine 80. J Exp Med 1994;180:1235–42. 10.1084/jem.180.4.1235
    1. Rojo S, Wagtmann N, Long EO. Binding of a soluble p70 killer cell inhibitory receptor to HLA-B*5101: requirement for all three p70 immunoglobulin domains. Eur J Immunol 1997;27:568–71. 10.1002/eji.1830270231
    1. Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol 2010;10:317–27. 10.1038/nri2744
    1. Kärre K, Ljunggren HG, Piontek G, et al. . Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature 1986;319:675–8. 10.1038/319675a0
    1. Ljunggren HG, Kärre K. In search of the 'missing self': MHC molecules and NK cell recognition. Immunol Today 1990;11:237–44. 10.1016/0167-5699(90)90097-S
    1. Pende D, Biassoni R, Cantoni C, et al. . The natural killer cell receptor specific for HLA-A allotypes: a novel member of the p58/p70 family of inhibitory receptors that is characterized by three immunoglobulin-like domains and is expressed as a 140-kD disulphide-linked dimer. J Exp Med 1996;184:505–18. 10.1084/jem.184.2.505
    1. Döhring C, Scheidegger D, Samaridis J, et al. . A human killer inhibitory receptor specific for HLA-A1,2. J Immunol 1996;156:3098–101.
    1. Hansasuta P, Dong T, Thananchai H, et al. . Recognition of HLA-A3 and HLA-A11 by KIR3DL2 is peptide-specific. Eur J Immunol 2004;34:1673–9. 10.1002/eji.200425089
    1. Yawata M, Yawata N, Draghi M, et al. . Mhc class I-specific inhibitory receptors and their ligands structure diverse human NK-cell repertoires toward a balance of missing self-response. Blood 2008;112:2369–80. 10.1182/blood-2008-03-143727
    1. Winter CC, Gumperz JE, Parham P, et al. . Direct binding and functional transfer of NK cell inhibitory receptors reveal novel patterns of HLA-C allotype recognition. J Immunol 1998;161:571–7.
    1. Babor F, Manser AR, Fischer JC, et al. . Kir ligand C2 is associated with increased susceptibility to childhood all and confers an elevated risk for late relapse. Blood 2014;124:2248–51. 10.1182/blood-2014-05-572065
    1. Giebel S, Locatelli F, Wojnar J, et al. . Homozygosity for human leucocyte antigen-C ligands of KIR2DL1 is associated with increased risk of relapse after human leucocyte antigen-C-matched unrelated donor haematopoietic stem cell transplantation. Br J Haematol 2005;131:483–6. 10.1111/j.1365-2141.2005.05797.x
    1. Gazit R, Garty B-Z, Monselise Y, et al. . Expression of KIR2DL1 on the entire NK cell population: a possible novel immunodeficiency syndrome. Blood 2004;103:1965–6. 10.1182/blood-2003-11-3796
    1. Boudreau JE, Giglio F, Gooley TA, et al. . KIR3DL1/HLA-B subtypes govern acute myelogenous leukemia relapse after hematopoietic cell transplantation. J Clin Oncol 2017;35:2268–78. 10.1200/JCO.2016.70.7059
    1. Hatjiharissi E, Xu L, Santos DD, et al. . Increased natural killer cell expression of CD16, augmented binding and ADCC activity to rituximab among individuals expressing the Fc{gamma}RIIIa-158 V/V and V/F polymorphism. Blood 2007;110:2561–4. 10.1182/blood-2007-01-070656
    1. Anderson K. Multiple myeloma: advances reported in 2013 are useful in the clinic. J Natl Compr Canc Netw 2014;12:808–11. 10.6004/jnccn.2014.0195
    1. Gluck WL, Hurst D, Yuen A, et al. . Phase I studies of interleukin (IL)-2 and rituximab in B-cell non-hodgkin's lymphoma: IL-2 mediated natural killer cell expansion correlations with clinical response. Clin Cancer Res 2004;10:2253–64. 10.1158/1078-0432.ccr-1087-3
    1. Benson DM, Cohen AD, Jagannath S, et al. . A phase I trial of the Anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma. Clin Cancer Res 2015;21:4055–61. 10.1158/1078-0432.CCR-15-0304
    1. Rajalingam R, Du Z, Meenagh A, et al. . Distinct diversity of KIR genes in three southern Indian populations: comparison with world populations revealed a link between Kir gene content and pre-historic human migrations. Immunogenetics 2008;60:207–17. 10.1007/s00251-008-0286-2

Source: PubMed

3
구독하다