Skip to main content

Advertisement

Log in

The CD38low natural killer cell line KHYG1 transiently expressing CD16F158V in combination with daratumumab targets multiple myeloma cells with minimal effector NK cell fratricide

  • Original Article
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Multiple myeloma (MM) is a clonal plasma cell malignancy typically associated with the high and uniform expression of the CD38 transmembrane glycoprotein. Daratumumab is a humanized IgG1κ CD38 monoclonal antibody (MoAb) which has demonstrated impressive single agent activity even in relapsed refractory MM patients as well as strong synergy with other anti-MM drugs. Natural Killer (NK) cells are cytotoxic immune effector cells that mediate in vivo tumour immunosurveillance. NK cells also play an important role during MoAb therapy by inducing antibody dependent cellular cytotoxicity (ADCC) via their FcγRIII (CD16) receptor. Furthermore, 15% of the population express a naturally occurring variant of CD16 harbouring a single-point polymorphism (F158V). However, the contribution of NK cells to the efficacy of daratumumab remains debatable as clinical data clearly indicate the rapid depletion of CD38high peripheral blood NK cells in patients upon daratumumab administration. In contrast, CD38low peripheral blood NK cells have been shown to survive daratumumab mediated fratricide in vivo, while still retaining their potent anti-MM cytolytic effector functions ex vivo. Therefore, we hypothesize that transiently expressing the CD16F158V receptor using a “safe” mRNA electroporation-based approach on CD38low NK cells in combination with daratumumab could represent a novel therapeutic option for treatment of MM. In the present study, we investigate a NK cell line (KHYG-1), derived from a patient with aggressive NK cell leukemia, as a platform for generating CD38low NK cells expressing CD16F158V which can be administered as an “off-the-shelf” therapy to target both CD38high and CD38low tumour clones in patients receiving daratumumab.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

ADCC:

Antibody dependent cellular cytotoxicity

ADCP:

Antibody dependent cellular phagocytosis

ADRP:

ADP-ribose pyrophosphate

CDC:

Complement-dependent cytotoxicity

E: T:

Effector: target

IMiDs:

Immunomodulatory drugs

KIR:

Killer inhibitory receptor

MM:

Multiple myeloma

MoAb:

Monoclonal antibody

NDMM:

Newly diagnosed multiple myeloma

NK:

Natural killer

PI:

Propidium iodide

References

  1. Sarkar S, Chauhan S, Stikvoort A, Natoni A, Daly J, Henderson R, Mutis T, O'Dwyer ME (2018) CD38(low) natural killer cells transiently expressing CD16(F158V) m-RNA potentiates the therapeutic activity of daratumumab against multiple myeloma with minimal effector NK cell fratricide. Blood. https://doi.org/10.1182/blood-2018-99-118926

    Article  Google Scholar 

  2. Palumbo A, Anderson K (2011) Multiple myeloma. N Engl J Med 364(11):1046–1060. https://doi.org/10.1056/NEJMra1011442

    Article  CAS  PubMed  Google Scholar 

  3. van de Donk N, Richardson PG, Malavasi F (2018) CD38 antibodies in multiple myeloma: back to the future. Blood 131(1):13–29. https://doi.org/10.1182/blood-2017-06-740944

    Article  CAS  PubMed  Google Scholar 

  4. van de Donk N, Yong K (2019) Oral proteasome inhibitor maintenance for multiple myeloma. Lancet 393(10168):204–205. https://doi.org/10.1016/S0140-6736(18)33128-3

    Article  PubMed  Google Scholar 

  5. Fast LD, Hansen JA, Newman W (1981) Evidence for T cell nature and heterogeneity within natural killer (NK) and antibody-dependent cellular cytotoxicity (ADCC) effectors: a comparison with cytolytic T lymphocytes (CTL). J Immunol 127(2):448–452

    CAS  PubMed  Google Scholar 

  6. Mahaweni NM, Bos GMJ, Mitsiades CS, Tilanus MGJ, Wieten L (2018) Daratumumab augments alloreactive natural killer cell cytotoxicity towards CD38+ multiple myeloma cell lines in a biochemical context mimicking tumour microenvironment conditions. Cancer Immunol Immunother 67(6):861–872. https://doi.org/10.1007/s00262-018-2140-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hatjiharissi E, Xu L, Santos DD, Hunter ZR, Ciccarelli BT, Verselis S, Modica M, Cao Y, Manning RJ, Leleu X, Dimmock EA, Kortsaris A, Mitsiades C, Anderson KC, Fox EA, Treon SP (2007) 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 110(7):2561–2564. https://doi.org/10.1182/blood-2007-01-070656

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Leo R, Boeker M, Peest D, Hein R, Bartl R, Gessner JE, Selbach J, Wacker G, Deicher H (1992) Multiparameter analyses of normal and malignant human plasma cells: CD38++, CD56+, CD54+, cIg+ is the common phenotype of myeloma cells. Ann Hematol 64(3):132–139

    Article  CAS  Google Scholar 

  9. Funaro A, Malavasi F (1999) Human CD38, a surface receptor, an enzyme, an adhesion molecule and not a simple marker. J Biol Regul Homeost Agents 13(1):54–61

    CAS  PubMed  Google Scholar 

  10. Palumbo A, Chanan-Khan A, Weisel K, Nooka AK, Masszi T, Beksac M, Spicka I, Hungria V, Munder M, Mateos MV, Mark TM, Qi M, Schecter J, Amin H, Qin X, Deraedt W, Ahmadi T, Spencer A, Sonneveld P, Investigators C (2016) Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med 375(8):754–766. https://doi.org/10.1056/NEJMoa1606038

    Article  CAS  PubMed  Google Scholar 

  11. Dimopoulos MA, San-Miguel J, Belch A, White D, Benboubker L, Cook G, Leiba M, Morton J, Ho PJ, Kim K, Takezako N, Moreau P, Kaufman JL, Sutherland HJ, Lalancette M, Magen H, Iida S, Kim JS, Prince HM, Cochrane T, Oriol A, Bahlis NJ, Chari A, O'Rourke L, Wu K, Schecter JM, Casneuf T, Chiu C, Soong D, Sasser AK, Khokhar NZ, Avet-Loiseau H, Usmani SZ (2018) Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POLLUX. Haematologica 103(12):2088–2096. https://doi.org/10.3324/haematol.2018.194282

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mateos MV, Dimopoulos MA, Cavo M, Suzuki K, Jakubowiak A, Knop S, Doyen C, Lucio P, Nagy Z, Kaplan P, Pour L, Cook M, Grosicki S, Crepaldi A, Liberati AM, Campbell P, Shelekhova T, Yoon SS, Iosava G, Fujisaki T, Garg M, Chiu C, Wang J, Carson R, Crist W, Deraedt W, Nguyen H, Qi M, San-Miguel J, Investigators AT (2018) Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med 378(6):518–528. https://doi.org/10.1056/NEJMoa1714678

    Article  CAS  PubMed  Google Scholar 

  13. Facon T, Kumar S, Plesner T, Orlowski RZ, Moreau P, Bahlis N, Basu S, Nahi H, Hulin C, Quach H, Goldschmidt H, O'Dwyer M, Perrot A, Venner CP, Weisel K, Mace JR, Raje N, Attal M, Tiab M, Macro M, Frenzel L, Leleu X, Ahmadi T, Chiu C, Wang J, Van Rampelbergh R, Uhlar CM, Kobos R, Qi M, Usmani SZ, Investigators MT (2019) Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med 380(22):2104–2115. https://doi.org/10.1056/NEJMoa1817249

    Article  CAS  PubMed  Google Scholar 

  14. van de Donk N, Usmani SZ (2018) CD38 antibodies in multiple myeloma: mechanisms of action and modes of resistance. Front Immunol 9:2134. https://doi.org/10.3389/fimmu.2018.02134

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Nijhof IS, Casneuf T, van Velzen J, van Kessel B, Axel AE, Syed K, Groen RW, van Duin M, Sonneveld P, Minnema MC, Zweegman S, Chiu C, Bloem AC, Mutis T, Lokhorst HM, Sasser AK, van de Donk NW (2016) CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma. Blood 128(7):959–970. https://doi.org/10.1182/blood-2016-03-703439

    Article  CAS  PubMed  Google Scholar 

  16. Rah SY, Kwak JY, Chung YJ, Kim UH (2015) ADP-ribose/TRPM2-mediated Ca2+ signaling is essential for cytolytic degranulation and antitumor activity of natural killer cells. Sci Rep 5:9482. https://doi.org/10.1038/srep09482

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Krejcik J, van de Donk N (2018) Trogocytosis represents a novel mechanism of action of daratumumab in multiple myeloma. Oncotarget 9(72):33621–33622. https://doi.org/10.18632/oncotarget.26098

    Article  PubMed  PubMed Central  Google Scholar 

  18. Usmani SZ, Khan I, Chiu C, Foureau D, Druhan LJ, Rigby K, Casneuf T, Sasser AK (2018) Deep sustained response to daratumumab monotherapy associated with T cell expansion in triple refractory myeloma. Exp Hematol Oncol 7:3. https://doi.org/10.1186/s40164-018-0096-7

    Article  PubMed  PubMed Central  Google Scholar 

  19. Wang Y, Zhang Y, Hughes T, Zhang J, Caligiuri MA, Benson DM, Yu J (2018) Fratricide of NK cells in daratumumab therapy for multiple myeloma overcome by ex vivo-expanded autologous NK cells. Clin Cancer Res 24(16):4006–4017. https://doi.org/10.1158/1078-0432.CCR-17-3117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Natoni A, Farrell ML, Harris S, Falank C, Kirkham-McCarthy L, Macauley MS, Reagan MR, O'Dwyer M (2019) Sialyltransferase inhibition leads to inhibition of tumor cell interactions with E-selectin, VCAM1, and MADCAM1, and improves survival in a human multiple myeloma mouse model. Haematologica. https://doi.org/10.3324/haematol.2018.212266

    Article  PubMed  Google Scholar 

  21. Srpan K, Ambrose A, Karampatzakis A, Saeed M, Cartwright ANR, Guldevall K, De Matos G, Onfelt B, Davis DM (2018) Shedding of CD16 disassembles the NK cell immune synapse and boosts serial engagement of target cells. J Cell Biol 217(9):3267–3283. https://doi.org/10.1083/jcb.201712085

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Warren HS, Kinnear BF (1999) Quantitative analysis of the effect of CD16 ligation on human NK cell proliferation. J Immunol 162(2):735–742

    CAS  PubMed  Google Scholar 

  23. Ponzetta A, Benigni G, Antonangeli F, Sciume G, Sanseviero E, Zingoni A, Ricciardi MR, Petrucci MT, Santoni A, Bernardini G (2015) Multiple myeloma impairs bone marrow localization of effector natural killer cells by altering the chemokine microenvironment. Cancer Res 75(22):4766–4777. https://doi.org/10.1158/0008-5472.CAN-15-1320

    Article  CAS  PubMed  Google Scholar 

  24. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, Minnema MC, Lassen U, Krejcik J, Palumbo A, van de Donk NW, Ahmadi T, Khan I, Uhlar CM, Wang J, Sasser AK, Losic N, Lisby S, Basse L, Brun N, Richardson PG (2015) Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med 373(13):1207–1219. https://doi.org/10.1056/NEJMoa1506348

    Article  CAS  PubMed  Google Scholar 

  25. Usmani SZ, Weiss BM, Plesner T, Bahlis NJ, Belch A, Lonial S, Lokhorst HM, Voorhees PM, Richardson PG, Chari A, Sasser AK, Axel A, Feng H, Uhlar CM, Wang J, Khan I, Ahmadi T, Nahi H (2016) Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood 128(1):37–44. https://doi.org/10.1182/blood-2016-03-705210

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Casneuf T, Xu XS, Adams HC 3rd, Axel AE, Chiu C, Khan I, Ahmadi T, Yan X, Lonial S, Plesner T, Lokhorst HM, van de Donk N, Clemens PL, Sasser AK (2017) Effects of daratumumab on natural killer cells and impact on clinical outcomes in relapsed or refractory multiple myeloma. Blood Adv 1(23):2105–2114. https://doi.org/10.1182/bloodadvances.2017006866

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nahi H, Chrobok M, Gran C, Lund J, Gruber A, Gahrton G, Ljungman P, Wagner AK, Alici E (2019) Infectious complications and NK cell depletion following daratumumab treatment of multiple myeloma. PLoS One 14(2):e0211927. https://doi.org/10.1371/journal.pone.0211927

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang W, Erbe AK, Hank JA, Morris ZS, Sondel PM (2015) NK cell-mediated antibody-dependent cellular cytotoxicity in cancer immunotherapy. Front Immunol 6:368. https://doi.org/10.3389/fimmu.2015.00368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fauriat C, Mallet F, Olive D, Costello RT (2006) Impaired activating receptor expression pattern in natural killer cells from patients with multiple myeloma. Leukemia 20(4):732–733. https://doi.org/10.1038/sj.leu.2404096

    Article  CAS  PubMed  Google Scholar 

  30. Sarkar S, Germeraad WT, Rouschop KM, Steeghs EM, van Gelder M, Bos GM, Wieten L (2013) Hypoxia induced impairment of NK cell cytotoxicity against multiple myeloma can be overcome by IL-2 activation of the NK cells. PLoS One 8(5):e64835. https://doi.org/10.1371/journal.pone.0064835

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard J 2nd, Wei M, Mao H, Byrd JC, Perrotti D, Caligiuri MA (2008) TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181(6):3784–3792. https://doi.org/10.4049/jimmunol.181.6.3784

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D, Luo X, Cooley S, Verneris M, Walcheck B, Miller J (2013) NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 121(18):3599–3608. https://doi.org/10.1182/blood-2012-04-425397

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Carlsten M, Levy E, Karambelkar A, Li L, Reger R, Berg M, Peshwa MV, Childs RW (2016) Efficient mRNA-based genetic engineering of human NK cells with high-affinity CD16 and CCR7 augments rituximab-induced ADCC against lymphoma and targets NK cell migration toward the lymph node-associated chemokine CCL19. Front Immunol 7:105. https://doi.org/10.3389/fimmu.2016.00105

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, MacIntyre E, Dal Cortivo L, Radford I, Brousse N, Sigaux F, Moshous D, Hauer J, Borkhardt A, Belohradsky BH, Wintergerst U, Velez MC, Leiva L, Sorensen R, Wulffraat N, Blanche S, Bushman FD, Fischer A, Cavazzana-Calvo M (2008) Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest 118(9):3132–3142. https://doi.org/10.1172/JCI35700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kobayashi E, Motoi S, Sugiura M, Kajikawa M, Kojima S, Kohroki J, Masuho Y (2014) Antibody-dependent cellular cytotoxicity and cytokine/chemokine secretion by KHYG-1 cells stably expressing FcgammaRIIIA. Immunol Lett 161(1):59–64. https://doi.org/10.1016/j.imlet.2014.05.004

    Article  CAS  PubMed  Google Scholar 

  36. Krejcik J, Frerichs KA, Nijhof IS, van Kessel B, van Velzen JF, Bloem AC, Broekmans MEC, Zweegman S, van Meerloo J, Musters RJP, Poddighe PJ, Groen RWJ, Chiu C, Plesner T, Lokhorst HM, Sasser AK, Mutis T, van de Donk N (2017) Monocytes and granulocytes reduce CD38 expression levels on myeloma cells in patients treated with daratumumab. Clin Cancer Res 23(24):7498–7511. https://doi.org/10.1158/1078-0432.CCR-17-2027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Weng WK, Levy R (2003) Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol 21(21):3940–3947. https://doi.org/10.1200/JCO.2003.05.013

    Article  CAS  PubMed  Google Scholar 

  38. van de Donk N, Casneuf T, Di Cara A, Parren PW, Zweegman S, van Kessel B, Lokhorst HM, Usmani SZ, Lonial S, Richardson PG, Chiu C, Mutis T, Nijhof IS, Sasser AK (2019) Impact of Fc gamma receptor polymorphisms on efficacy and safety of daratumumab in relapsed/refractory multiple myeloma. Br J Haematol 184(3):475–479. https://doi.org/10.1111/bjh.15122

    Article  PubMed  Google Scholar 

  39. Nijhof IS, Lammerts van Bueren JJ, van Kessel B, Andre P, Morel Y, Lokhorst HM, van de Donk NW, Parren PW, Mutis T (2015) Daratumumab-mediated lysis of primary multiple myeloma cells is enhanced in combination with the human anti-KIR antibody IPH2102 and lenalidomide. Haematologica 100(2):263–268. https://doi.org/10.3324/haematol.2014.117531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Garcia-Guerrero E, Gogishvili T, Danhof S, Schreder M, Pallaud C, Perez-Simon JA, Einsele H, Hudecek M (2017) Panobinostat induces CD38 upregulation and augments the antimyeloma efficacy of daratumumab. Blood 129(25):3386–3388. https://doi.org/10.1182/blood-2017-03-770776

    Article  CAS  PubMed  Google Scholar 

  41. Nijhof IS, Groen RW, Lokhorst HM, van Kessel B, Bloem AC, van Velzen J, de Jong-Korlaar R, Yuan H, Noort WA, Klein SK, Martens AC, Doshi P, Sasser K, Mutis T, van de Donk NW (2015) Upregulation of CD38 expression on multiple myeloma cells by all-trans retinoic acid improves the efficacy of daratumumab. Leukemia 29(10):2039–2049. https://doi.org/10.1038/leu.2015.123

    Article  CAS  PubMed  Google Scholar 

  42. Wang R, Jaw JJ, Stutzman NC, Zou Z, Sun PD (2012) Natural killer cell-produced IFN-gamma and TNF-alpha induce target cell cytolysis through up-regulation of ICAM-1. J Leukoc Biol 91(2):299–309. https://doi.org/10.1189/jlb.0611308

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ausiello CM, Urbani F, la Sala A, Funaro A, Malavasi F (1995) CD38 ligation induces discrete cytokine mRNA expression in human cultured lymphocytes. Eur J Immunol 25(5):1477–1480. https://doi.org/10.1002/eji.1830250554

    Article  CAS  PubMed  Google Scholar 

  44. Grzywacz B, Moench L, McKenna D Jr, Tessier KM, Bachanova V, Cooley S, Miller JS, Courville EL (2019) Natural killer cell homing and persistence in the bone marrow after adoptive immunotherapy correlates with better leukemia control. J Immunother 42(2):65–72. https://doi.org/10.1097/CJI.0000000000000250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Levy E, Reger R, Segerberg F, Lambert M, Leijonhufvud C, Baumer Y, Carlsten M, Childs R (2019) Enhanced bone marrow homing of natural killer cells following mRNA transfection with gain-of-function variant CXCR4(R334X). Front Immunol 10:1262. https://doi.org/10.3389/fimmu.2019.01262

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sconocchia G, Titus JA, Mazzoni A, Visintin A, Pericle F, Hicks SW, Malavasi F, Segal DM (1999) CD38 triggers cytotoxic responses in activated human natural killer cells. Blood 94(11):3864–3871

    Article  CAS  Google Scholar 

  47. Suck G, Branch DR, Smyth MJ, Miller RG, Vergidis J, Fahim S, Keating A (2005) KHYG-1, a model for the study of enhanced natural killer cell cytotoxicity. Exp Hematol 33(10):1160–1171. https://doi.org/10.1016/j.exphem.2005.06.024

    Article  CAS  PubMed  Google Scholar 

  48. Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, Syed K, Liu K, van de Donk NW, Weiss BM, Ahmadi T, Lokhorst HM, Mutis T, Sasser AK (2016) Daratumumab depletes CD38+ immune regulatory cells, promotes T cell expansion, and skews T cell repertoire in multiple myeloma. Blood 128(3):384–394. https://doi.org/10.1182/blood-2015-12-687749

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pick M, Vainstein V, Goldschmidt N, Lavie D, Libster D, Gural A, Grisariu S, Avni B, Ben Yehuda D, Gatt ME (2018) Daratumumab resistance is frequent in advanced-stage multiple myeloma patients irrespective of CD38 expression and is related to dismal prognosis. Eur J Haematol 100(5):494–501. https://doi.org/10.1111/ejh.13046

    Article  CAS  PubMed  Google Scholar 

  50. Minarik J, Novak M, Flodr P, Balcarkova J, Mlynarcikova M, Krhovska P, Pika T, Pikalova Z, Bacovsky J, Scudla V (2017) CD38-negative relapse in multiple myeloma after daratumumab-based chemotherapy. Eur J Haematol 99(2):186–189. https://doi.org/10.1111/ejh.12902

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Flowcytometry Core Facility at National University of Ireland Galway (NUIG), Ireland.

Funding

This research was supported by a Sponsored Research Agreement from ONK Therapeutics Ltd., Ireland to National University of Ireland Galway. Subhashis Sarkar received a Post-doctoral Fellowship from ONK Therapeutics Ltd. Michaela R. Reagan is supported in part by a Research Scholar Grant, RSG-19-037-01-LIB from the American Cancer Society, and the NIH, P20GM121301.

Author information

Authors and Affiliations

Authors

Contributions

SS, SKSC, JD, HF and MRR performed experiments. SS, SKSC, JD and HF analysed data. AN, RH, EN, DS, JH, and MRR assisted with experiments. SS and MOD designed the project. SS and MOD wrote the manuscript.

Corresponding author

Correspondence to Michael O’Dwyer.

Ethics declarations

Conflict of interest

Subhashis Sarkar: ONK Therapeutics Ltd.: Research funding; Michael O'Dwyer: Abbvie: Membership on advisory committee; Celgene: Membership on advisory committee, and research funding; Bristol Myers Squibb (BMS): Research funding; Glycomimetics: Research funding; ONK Therapeutics Ltd.: Equity Ownership, Membership on Board of Directors, and research funding; Janssen: Membership on advisory committees, and research funding; Michaela R. Reagan: ONK Therapeutics Ltd.: Research funding. A provisional patent application was filed by Subhashis Sarkar and Michael O'Dwyer regarding the CD16 work described in the paper. All other authors declare no conflict of interest.

Ethical approval

All experiments utilizing primary cells from MM patients were conducted in accordance with the ethical standards of the Clinical Research Ethics Committee University Hospital Galway and the 1964 Declaration of Helsinki. The collection of bone marrow samples from MM patients was approved by Clinical Research Ethics Committee University Hospital Galway on June 14, 2017 (Study no. C.A.1519) All experiments utilizing blood samples from healthy donors were conducted in accordance with the ethical standards of the Clinical Research Ethics Committee University Hospital Galway and the 1964 Declaration of Helsinki. The collection of blood samples from healthy volunteers was approved by Clinical Research Ethics Committee University Hospital Galway on August 4, 2017 (Study no. C.A.1805) Animal research experiments to examine in vivo bone marrow homing were performed at the Maine Medical Center Research Institute with approval from the institutional animal care and use committee (IACUC); (Protocol #MR1508), an (Association for assessment and accrediation of laboratory animal care) AAALAC international accredited program. Approval for the experiments in this manuscript was issued by the Maine Medical Center Research Institute (MMCRI) Institutional Animal care and use committee (IACUC) on September 2, 2016.

Informed consent

NK cells were freshly isolated from peripheral blood samples of anonymous volunteer healthy donors at the University Hospital Galway, Ireland. Oral informed consent was obtained from all blood donors to the use of their blood for scientific purposes. Bone Marrow samples were provided by the Haematology Unit of University Hospital Galway, Galway from MM patients who had provided written consent for remaining samples to be used for immunology related research.

Animal source

Female NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice (Stock# 005557) were purchased at 4-weeks of age from Jackson Laboratory (Bar Harbor, ME) and acclimated at the Maine Medical Center Research Institute animal facility for at least two weeks prior being randomly assigned to each group for experiments.

Cell line authentication

KHYG1 was a kind gift from Dr. Armand Keating, University of Toronto (Canada) and the cell line was authenticated by STR profiling on 06/06/2017. MM1S, RPMI-8226, JJN3, H929, and U266, were obtained from American Type Culture Collection (ATCC) (Manassas, VA, USA). NK-92 was purchased from Deutsche Sammlung von Mikcroorganismen and Zellkulturen  (DSMZ) (Braunschweig, Germany) The cell lines were initially grown and cryopreserved into multiple aliquots of master cell bank (MCB). All the experiments were performed with cells at low passage numbers (≤ 10). Mycoplasma was routinely tested.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Note on previous publication: Part of this work was presented as a poster at the annual conference of the American Society of Haematology (ASH) 1–4 December 2018, San Diego, USA (Session-Myeloma: Pathophysiology and Pre-Clinical Studies, excluding Therapy: Poster II). The accompanying abstract was published in a special edition of Blood [1].

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sarkar, S., Chauhan, S.K.S., Daly, J. et al. The CD38low natural killer cell line KHYG1 transiently expressing CD16F158V in combination with daratumumab targets multiple myeloma cells with minimal effector NK cell fratricide. Cancer Immunol Immunother 69, 421–434 (2020). https://doi.org/10.1007/s00262-019-02477-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-019-02477-8

Keywords

Navigation