Abstract
Multiple myeloma (MM) remains an incurable disease and there is an unmet medical need for novel therapeutic drugs that do not share similar mechanisms of action with currently available agents. Sphingosine kinase 2 (SK2) is an innovative molecular target for anticancer therapy. We previously reported that treatment with SK2 inhibitor opaganib inhibited myeloma tumor growth in vitro and in vivo in a mouse xenograft model. In the current study, we performed a phase I study of opaganib in patients with relapsed/refractory multiple myeloma (RRMM). Thirteen patients with RRMM previously treated with immunomodulatory agents and proteasome inhibitors were enrolled and treated with single-agent opaganib at three oral dosing regimens (250 mg BID, 500 mg BID, or 750 mg BID, 28 days as a cycle). Safety and maximal tolerated dose (MTD) were determined. Pharmacokinetics, pharmacodynamics, and correlative studies were also performed. Opaganib was well tolerated up to a dose of 750 mg BID. The most common possibly related adverse event (AE) was decreased neutrophil counts. There were no serious AEs considered to be related to opaganib. MTD was determined as at least 750 mg BID. On an intent-to-treat basis, one patient (7.7%) in the 500 mg BID dose cohort showed a very good partial response, and one other patient (7.7%) achieved stable disease for 3 months. SK2 is an innovative molecular target for antimyeloma therapy. The first-in-class SK2 inhibitor opaganib is generally safe for administration to RRMM patients, and has potential therapeutic activity in these patients. Clinicaltrials.gov: NCT02757326.
This is a preview of subscription content, access via your institution.


Data availability
The data that support the findings of this study are available on request from the corresponding author. Certain data are not publicly available due to privacy or ethical restrictions.
Abbreviations
- AE:
-
Adverse event
- BID:
-
Twice a day
- CAR:
-
Chimeric antigen receptor
- CTCAE:
-
Common Terminology Criteria for Adverse Events
- DCEP:
-
Dexamethasone, cyclophosphamide, etoposide, and cisplatin
- DLT:
-
Dose-limiting toxicity
- ECOG PS:
-
Eastern Cooperative Oncology Group performance status
- IMiD:
-
Immunomodulatory drug
- IMWG:
-
International Myeloma Working Group
- MM:
-
Multiple myeloma
- MTD:
-
Maximal tolerated dose
- NDMM:
-
Newly diagnosed multiple myeloma
- PD:
-
Pharmacodynamic
- PFS:
-
Progression-free survival
- PI:
-
Proteasome inhibitor
- PK:
-
Pharmacokinetic
- PrD:
-
Progressive disease
- RRMM:
-
Relapsed/refractory multiple myeloma
- SAE:
-
Serious adverse event
- SD:
-
Stable disease
- SK:
-
Sphingosine kinase
- VD-PACE:
-
Velcade, dexamethasone, cisplatin, doxorubicin, cyclophosphamide, and etoposide
- VGPR:
-
Very good partial response
References
Siegel R, Miller K, Fuchs H (2021) Cancer statistics, 2021. CA Cancer J Clin 71:7–33
Callander NS, Baljevic M, Adekola K, Anderson LD, Campagnaro E, Castillo JJ, Costello C, Devarakonda S, Elsedawy N, Faiman M et al (2022) NCCN Guidelines® Insights: Multiple Myeloma, Version 3.2022. J Natl Compr Canc Netw 20(1):8–19
Feinberg D, Paul B, Kang Y (2019) The promise of chimeric antigen receptor (CAR) T cell therapy in multiple myeloma. Cellular Immunol 345:103964
Usmani SZ, Garfall AL, van de Donk N, Nahi H, San-Miguel JF, Oriol A, Rosinol L, Chari A, Bhutani M, Karlin L et al (2021) Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): a multicentre, open-label, single-arm, phase 1 study. Lancet 398(10301):665–674
Yu B, Jiang T, Liu D (2020) BCMA-targeted immunotherapy for multiple myeloma. J Hematol Oncol 13(1):125
Moreau P, Garfall AL, van de Donk N, Nahi H, San-Miguel JF, Oriol A, Nooka AK, Martin T, Rosinol L, Chari A et al (2022) Teclistamab in relapsed or refractory multiple myeloma. N Engl J Med 387(6):495–505
Perrot A, Lauwers-Cances V, Cazaubiel T, Facon T, Caillot D, Clement-Filliatre L, Macro M, Decaux O, Belhadj K, Mohty M et al (2020) Early versus late autologous stem cell transplant in newly diagnosed multiple myeloma: long-term follow-up analysis of the IFM 2009 Trial. Blood 136(Supplement 1):39–39
Moreau P, Attal M, Hulin C, Arnulf B, Belhadj K, Benboubker L, Béné MC, Broijl A, Caillon H, Caillot D et al (2019) Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 394(10192):29–38
Lewis CS, Voelkel-Johnson C, Smith CD (2018) Targeting sphingosine kinases for the treatment of cancer. Adv Cancer Res 140:295–325
Perry DK, Kolesnick RN (2003) Ceramide and sphingosine 1-phosphate in anti-cancer therapies. Cancer Treat Res 115:345–354
Spiegel S, Milstien S (2002) Sphingosine 1-phosphate, a key cell signaling molecule. J Biol Chem 277(29):25851–25854
Gupta P, Taiyab A, Hussain A, Alajmi MF, Islam A, Hassan MI (2021) Targeting the sphingosine kinase/sphingosine-1-phosphate signaling axis in drug discovery for cancer therapy. Cancers (Basel) 13(8):1898
Hait NC, Maiti A (2017) The role of sphingosine-1-phosphate and ceramide-1-phosphate in inflammation and cancer. Mediators Inflamm 2017:4806541
Ogretmen B (2018) Sphingolipid metabolism in cancer signalling and therapy. Nat Rev Cancer 18(1):33–50
Kolesnick R, Fuks Z (2003) Radiation and ceramide-induced apoptosis. Oncogene 22(37):5897–5906
Pl T, Wang HG (2011) Editorial [hot topic: therapeutic targeting of the sphingolipid “biostat” in hematologic malignancies (guest editors: Thomas p. Loughran and Hong-gang wang)]. Anticancer Agents Med Chem 11(9):780–781
Leong WI, Saba JD (2010) S1P metabolism in cancer and other pathological conditions. Biochimie 92(6):716–723
Snider AJ, Alexa Orr Gandy K, Obeid LM (2010) Sphingosine kinase: role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie 92(6):707–715
Furuya H, Shimizu Y, Kawamori T (2011) Sphingolipids in cancer. Cancer Metastasis Rev 30(3–4):567–576
Santos CR, Schulze A (2012) Lipid metabolism in cancer. FEBS J 279(15):2610–2623
Obinata H, Hla T (2019) Sphingosine 1-phosphate and inflammation. Int Immunol 31(9):617–625
Venkata JK, An N, Stuart R, Costa LJ, Cai H, Coker W, Song JH, Gibbs K, Matson T, Garrett-Mayer E et al (2014) Inhibition of sphingosine kinase 2 downregulates the expression of c-Myc and Mcl-1 and induces apoptosis in multiple myeloma. Blood 124(12):1915–1925
French KJ, Zhuang Y, Maines LW, Gao P, Wang W, Beljanski V, Upson JJ, Green CL, Keller SN, Smith CD (2010) Pharmacology and antitumor activity of ABC294640, a selective inhibitor of sphingosine kinase-2. J Pharmacol Exp Ther 333(1):129–139
Sundaramoorthy P, Gasparetto C, Kang Y (2018) The combination of a sphingosine kinase 2 inhibitor (ABC294640) and a Bcl-2 inhibitor (ABT-199) displays synergistic anti-myeloma effects in myeloma cells without a t(11;14) translocation. Cancer Med 7(7):3257–3268
Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, Munshi N, Lonial S, Blade J, Mateos MV et al (2016) International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol 17(8):e328–e346
Kyle RA, Rajkumar SV (2009) Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia 23(1):3–9
Britten CD, Garrett-Mayer E, Chin SH, Shirai K, Ogretmen B, Bentz TA, Brisendine A, Anderton K, Cusack SL, Maines LW et al (2017) A phase i study of ABC294640, a first-in-class sphingosine kinase-2 inhibitor, in patients with advanced solid tumors. Clin Cancer Res 23(16):4642–4650
Hammad SM, Pierce JS, Soodavar F, Smith KJ, Al Gadban MM, Rembiesa B, Klein RL, Hannun YA, Bielawski J, Bielawska A (2010) Blood sphingolipidomics in healthy humans: impact of sample collection methodology. J Lipid Res 51(10):3074–3087
Daimon T, Zohar S, O’Quigley J (2011) Posterior maximization and averaging for Bayesian working model choice in the continual reassessment method. Stat Med 30(13):1563–1573
Yuan Y, Yin G (2011) Robust EM continual reassessment method in oncology dose finding. J Am Stat Assoc 106(495):818–831
Yin G, Yuan Y (2009) Bayesian model averaging continual reassessment method in phase I clinical trial. J Am Stat Assoc 104:954–968
Liu Q, Rehman H, Shi Y, Krishnasamy Y, Lemasters JJ, Smith CD, Zhong Z (2012) Inhibition of sphingosine kinase-2 suppresses inflammation and attenuates graft injury after liver transplantation in rats. PLoS ONE 7(7):e41834
Fitzpatrick LR, Green C, Frauenhoffer EE, French KJ, Zhuang Y, Maines LW, Upson JJ, Paul E, Donahue H, Mosher TJ et al (2011) Attenuation of arthritis in rodents by a novel orally-available inhibitor of sphingosine kinase. Inflammopharmacol 19(2):75–87
Maines LW, Fitzpatrick LR, Green CL, Zhuang Y, Smith CD (2010) Efficacy of a novel sphingosine kinase inhibitor in experimental Crohn’s disease. Inflammopharmacol 18(2):73–85
Pei G, Zyla J, He L, Moura-Alves P, Steinle H, Saikali P, Lozza L, Nieuwenhuizen N, Weiner J, Mollenkopf HJ et al (2021) Cellular stress promotes NOD1/2-dependent inflammation via the endogenous metabolite sphingosine-1-phosphate. Embo j 40(13):e106272
Gomez-Larrauri A, Presa N, Dominguez-Herrera A, Ouro A, Trueba M, Gomez-Muñoz A (2020) Role of bioactive sphingolipids in physiology and pathology. Essays Biochem 64(3):579–589
Hannun YA, Obeid LM (2018) Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol 19(3):175–191
Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S, Spiegel S (1996) Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature 381(6585):800–803
Olivera A, Spiegel S (2001) Sphingosine kinase: a mediator of vital cellular functions. Prostaglandins 64(1–4):123–134
Cowart LA (2009) Sphingolipids: players in the pathology of metabolic disease. Trends Endocrinol Metab 20(1):34–42
Saddoughi SA, Song P, Ogretmen B (2008) Roles of bioactive sphingolipids in cancer biology and therapeutics. Subcell Biochem 49:413–440
Cuvillier O (2002) Sphingosine in apoptosis signaling. Biochim Biophys Acta 1585(2–3):153–162
Leung RW, Alison JA, McKeough ZJ, Peters MJ (2011) A study design to investigate the effect of short-form Sun-style Tai Chi in improving functional exercise capacity, physical performance, balance and health related quality of life in people with chronic obstructive pulmonary disease (COPD). Contemp Clin Trials 32(2):267–272
Lee CF, Dang A, Hernandez E, Pong RC, Chen B, Sonavane R, Raj G, Kapur P, Lin HY, Wu SR et al (2019) Activation of sphingosine kinase by lipopolysaccharide promotes prostate cancer cell invasion and metastasis via SphK1/S1PR4/matriptase. Oncogene 38(28):5580–5598
Nagahashi M, Yamada A, Katsuta E, Aoyagi T, Huang WC, Terracina KP, Hait NC, Allegood JC, Tsuchida J, Yuza K et al (2018) Targeting the SphK1/S1P/S1PR1 axis that links obesity, chronic inflammation, and breast cancer metastasis. Cancer Res 78(7):1713–1725
Ader I, Brizuela L, Bouquerel P, Malavaud B, Cuvillier O (2008) Sphingosine kinase 1: a new modulator of hypoxia inducible factor 1alpha during hypoxia in human cancer cells. Cancer Res 68(20):8635–8642
Ahmad M, Long JS, Pyne NJ, Pyne S (2006) The effect of hypoxia on lipid phosphate receptor and sphingosine kinase expression and mitogen-activated protein kinase signaling in human pulmonary smooth muscle cells. Prostaglandins Other Lipid Mediat 79(3–4):278–286
Anelli V, Gault CR, Cheng AB, Obeid LM (2008) Sphingosine kinase 1 is up-regulated during hypoxia in U87MG glioma cells. Role of hypoxia-inducible factors 1 and 2. J Biol Chem 283(6):3365–3375
Schwalm S, Doll F, Romer I, Bubnova S, Pfeilschifter J, Huwiler A (2008) Sphingosine kinase-1 is a hypoxia-regulated gene that stimulates migration of human endothelial cells. Biochem Biophys Res Commun 368(4):1020–1025
Acharya S, Yao J, Li P, Zhang C, Lowery FJ, Zhang Q, Guo H, Qu J, Yang F, Wistuba II et al (2019) Sphingosine kinase 1 signaling promotes metastasis of triple-negative breast cancer. Cancer Res 79(16):4211–4226
Heffernan-Stroud LA, Obeid LM (2013) Sphingosine kinase 1 in cancer. Adv Cancer Res 117:201–235
Cuvillier O, Ader I, Bouquerel P, Brizuela L, Malavaud B, Mazerolles C, Rischmann P (2010) Activation of sphingosine kinase-1 in cancer: implications for therapeutic targeting. Curr Mol Pharmacol 3(2):53–65
Snider AJ, Orr Gandy KA, Obeid LM (2010) Sphingosine kinase: role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie 92(6):707–715
Yuza K, Nakajima M, Nagahashi M, Tsuchida J, Hirose Y, Miura K, Tajima Y, Abe M, Sakimura K, Takabe K et al (2018) Different roles of sphingosine kinase 1 and 2 in pancreatic cancer progression. J Surg Res 232:186–194
Kunkel GT, Maceyka M, Milstien S, Spiegel S (2013) Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond. Nat Rev Drug Discov 12(9):688–702
Pyne NJ, Adams DR, Pyne S (2017) Sphingosine kinase 2 in autoimmune/inflammatory disease and the development of sphingosine kinase 2 inhibitors. Trends Pharmacol Sci 38(7):581–591
Song DD, Zhou JH, Sheng R (2018) Regulation and function of sphingosine kinase 2 in diseases. Histol Histopathol 33(5):433–445
van Echten-Deckert G, Alam S (2018) Sphingolipid metabolism - an ambiguous regulator of autophagy in the brain. Biol Chem 399(8):837–850
Jafari N, Drury J, Morris AJ, Onono FO, Stevens PD, Gao T, Liu J, Wang C, Lee EY, Weiss HL et al (2019) De novo fatty acid synthesis-driven sphingolipid metabolism promotes metastatic potential of colorectal cancer. Mol Cancer Res 17(1):140–152
Gao P, Smith CD (2011) Ablation of sphingosine kinase-2 inhibits tumor cell proliferation and migration. Mol Cancer Res 9(11):1509–1519
Acknowledgements
We thank patients and their families for their support and participation in this study.
Funding
This work was supported by NCI R44CA199767 (SBIR award to CS), NCI R01CA197792 (YK), NCI R21CA234701 (YK), NCI P01CA203628 (BO) and Duke Cancer Center Start-up fund (YK). RedHill Biopharma Limited is the IND holder and sponsored the trial including data monitoring and CRO activities.
Author information
Authors and Affiliations
Contributions
YK: designed the experiments and wrote the manuscript; PS, DF, and SF: performed the correlative studies; CG, GL, ES, AG, SAT, and BNR: accrued patients for the studies; ZL: provided biostatistical support; BL and BO: provided important inputs; LM and VKB-Y: supported the clinical studies; CS: designed the clinical study, wrote and edited the manuscript, and the awardee of the NCI SBIR grant; TP: Medical Director for RedHill Biopharma, the sponsor of the clinical study, wrote and edited the manuscript, and provided input on the protocol and analysis of results.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was conducted according to the Declaration of Helsinki and the International Council on Harmonization. The Duke Institutional Review Board (IRB) approved the protocol. All patients provided written informed consent.
Consent for publication
All authors approved the manuscript and the submission.
Competing interests
Yubin Kang received research funding from InCyte Corporation and a consultancy fee from Takeda Oncology USA and Sanofi Genzyme Corp. Charles Smith is the CEO and President, and Lynn Maines is employed by Apogee Biotechnology Corporation. Vered Katz Ben-Yair and Terry Plasse are consultants to RedHill Biopharma Limited. All other authors declare no competing conflicts of interest.
Additional information
Publisher's note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Below is the link to the electronic supplementary material.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Kang, Y., Sundaramoorthy, P., Gasparetto, C. et al. Phase I study of opaganib, an oral sphingosine kinase 2-specific inhibitor, in relapsed and/or refractory multiple myeloma. Ann Hematol 102, 369–383 (2023). https://doi.org/10.1007/s00277-022-05056-7
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1007/s00277-022-05056-7
Keywords
- Phase I trial
- Multiple myeloma
- Opaganib
- ABC294640
- Sphingosine kinase inhibitor
- Maximal tolerated dose
- Response