Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours

David S Hong, Yoon-Koo Kang, Mitesh Borad, Jasgit Sachdev, Samuel Ejadi, Ho Yeong Lim, Andrew J Brenner, Keunchil Park, Jae-Lyun Lee, Tae-You Kim, Sangjoon Shin, Carlos R Becerra, Gerald Falchook, Jay Stoudemire, Desiree Martin, Kevin Kelnar, Heidi Peltier, Vinicius Bonato, Andreas G Bader, Susan Smith, Sinil Kim, Vincent O'Neill, Muhammad S Beg, David S Hong, Yoon-Koo Kang, Mitesh Borad, Jasgit Sachdev, Samuel Ejadi, Ho Yeong Lim, Andrew J Brenner, Keunchil Park, Jae-Lyun Lee, Tae-You Kim, Sangjoon Shin, Carlos R Becerra, Gerald Falchook, Jay Stoudemire, Desiree Martin, Kevin Kelnar, Heidi Peltier, Vinicius Bonato, Andreas G Bader, Susan Smith, Sinil Kim, Vincent O'Neill, Muhammad S Beg

Abstract

Background: In this first-in-human, Phase 1 study of a microRNA-based cancer therapy, the recommended Phase 2 dose (RP2D) of MRX34, a liposomal mimic of microRNA-34a (miR-34a), was determined and evaluated in patients with advanced solid tumours.

Methods: Adults with various solid tumours refractory to standard treatments were enrolled in 3 + 3 dose-escalation cohorts and, following RP2D determination, expansion cohorts. MRX34, with oral dexamethasone premedication, was given intravenously daily for 5 days in 3-week cycles.

Results: Common all-cause adverse events observed in 85 patients enrolled included fever (% all grade/G3: 72/4), chills (53/14), fatigue (51/9), back/neck pain (36/5), nausea (36/1) and dyspnoea (25/4). The RP2D was 70 mg/m2 for hepatocellular carcinoma (HCC) and 93 mg/m2 for non-HCC cancers. Pharmacodynamic results showed delivery of miR-34a to tumours, and dose-dependent modulation of target gene expression in white blood cells. Three patients had PRs and 16 had SD lasting ≥4 cycles (median, 19 weeks, range, 11-55).

Conclusion: MRX34 treatment with dexamethasone premedication demonstrated a manageable toxicity profile in most patients and some clinical activity. Although the trial was closed early due to serious immune-mediated AEs that resulted in four patient deaths, dose-dependent modulation of relevant target genes provides proof-of-concept for miRNA-based cancer therapy.

Clinical trial registration: NCT01829971.

Conflict of interest statement

D.S.H. Research/Grant Funding: AbbVie, Adaptimmune, Amgen, Astra-Zeneca, Bayer, BMS, Daiichi-Sankyo, Eisai, Fate Therapeutics, Genentech, Genmab, Ignyta, Infinity, Kite, Kyowa, Lilly, LOXO, Merck, MedImmune, Mirati, MiRNA, Molecular Templates, Mologen, NCI-CTEP, Novartis, Pfizer, Seattle Genetics, Takeda Travel, Accommodations, Expenses: LOXO, MiRNA; Consulting or advisory role: Alpha Insights, Axiom, Adaptimmune, Baxter, Bayer (Ad Board and Speakers Bureau), Genentech, GLG, Group H, Guidepoint Global, Infinity, Janssen, Merrimack, Medscape, Numab, Pfizer, Seattle Genetics, Takeda, Trieza Therapeutics other ownership interests: Molecular Match (Advisor), OncoResponse (founder), Presagia Inc (Advisor). Y.-K.K. consulting or advisory role: Lilly/ImClone; Novartis; Ono Pharmaceutical; Roche/ Genentech; Taiho Pharmaceutical; research funding: Bayer; Novartis; Roche/Genentech. J.Sa. Honoraria: Celgene; consulting or Advisory Role: Celgene. A.B. Honoraria: Vascular Biogenics; Consulting or Advisory Role: NanoTX; Teleflex Medical Research Funding: Mirna Therapeutics (Inst); Threshold Pharmaceuticals; Patents, Royalties, other intellectual property: NanoTx Pharmaceuticals; travel, accommodations, expenses: Vascular Biogenics. G.F. Royalties: Wolters Kluwer; Advisory role: EMD Serono; Travel: Bristol-Myers Squibb, EMD Serono, Millennium; Research funding: 3-V Biosciences, Abbvie, Aileron, American Society of Clinical Oncology, Amgen, ARMO, AstraZeneca, BeiGene, Biothera, Celldex, Celgene, Ciclomed, Curegenix, Curis, DelMar, eFFECTOR, Eli Lilly, EMD Serono, Fujifilm, Genmab, GlaxoSmithKline, Hutchison MediPharma, Ignyta, Incyte, Jacobio, Jounce, Kolltan, Loxo, MedImmune, Millennium, Merck, miRNA Therapeutics, National Institutes of Health, Novartis, OncoMed, Oncothyreon, Precision Oncology, Regeneron, Rgenix, Strategia, Syndax, Taiho, Takeda, Tarveda, Tesaro, Tocagen, U.T. MD Anderson Cancer Center, Vegenics. A.G.B. employment: Mirna Therapeutics; stock and other ownership interests: Mirna Therapeutics Patents, royalties, other intellectual property: inventor on patents and patent applications assigned to Mirna Therapeutics. J.St. employment: Mirna Therapeutics; stock and other ownership interests: Mirna Therapeutics. S.S. employment: Mirna Therapeutics; stock and other ownership interests: Mirna Therapeutics. S.K. employment: Mirna Therapeutics; Leadership: Mirna Therapeutics; stock and other ownership interests: Mirna Therapeutics; Pfizer; Patents, Royalties, other intellectual property: listed as an inventor on patent applications, but no ownership interest or royalties. M.S.B. consulting or advisory role: Bayer; Celgene; Ipsen; Research funding: Celgene; Mirna Therapeutics; Precision Biologics; Travel, Accommodations, Expenses: Mirna Therapeutics; Precision Biologics. The remaining authors declare no competing interests.

Figures

Fig. 1. MRX34 pharmacodynamics.
Fig. 1. MRX34 pharmacodynamics.
a As measured by qRT-PCR, combined relative mRNA expression (%) pre- and post-treatment (5 timepoints) of five miR-34a target oncogenes (BCL2, CTNNB1, DNAJB1, FOXP1, HDAC1) in white blood cells (WBCs) from patients shows dose-dependent downregulation with increasing MRX34 dose from 50 (n = 4) to 70 (n = 16) to 93 (n = 16) to 110 mg/m2 (n = 9). Average expression in pre-dose samples was set as 100%. b Next generation sequencing (RNA-Seq) measurement of relative mRNA expression (%) pre- and post-treatment (24 h) for validated miR-34a target genes shows similar results to qRT-PCR. Dose-dependent knockdown is suggested by the increased range of downregulated genes seen with increasing MRX34 dose from 50 (n = 4) to 70 (n = 6) to 93 (n = 9) to 110 mg/m2 (n = 11). Validated miR-34a target genes contain miR-34a binding sites in their respective 3′ UTRs for which regulation by miR-34a has been experimentally verified. c Sylamer analysis plot shows a statistically significant enriched miRNA signature for miR-34a for putative target genes, suggesting specific activity of the miR-34a mimic against its target genes in patient-derived WBCs. In this analysis, genes affected by MRX34 at the 24-hr time point relative to baseline in the pre-dose samples for all dosing cohorts were sorted based on RNA-Seq measured expression from most downregulated to most upregulated. The 3′ UTR sequences of these genes were then scanned by the Sylamer algorithm for miRNA binding sites. The x-axis of the plot represents the sorted gene list from downregulated to upregulated. The y-axis represents an enrichment score of the seed sequence binding sites. P-values are based on simulations for seed ‘CTGCCA’. The plot also includes results of Sylamer analyses performed for house-keeping miRNAs, miR-24 and miR-191, which failed to show significant enrichment of downregulated target genes for these miRNAs, further indicating specific gene-directed activity of MRX34.
Fig. 2. Chromogenic in situ hybridisation (CISH)…
Fig. 2. Chromogenic in situ hybridisation (CISH) staining of pre- (baseline) and post-MRX34 treatment (treated) liver biopsies from patients with various advanced solid tumours.
Compared to pre-treatment staining, post-treatment results show variably increased miR-34a staining (dark blue/purple) in tumour tissue with localisation to the cellular cytoplasm. In all cases, a lesion in the liver was biopsied for CISH analysis. Uveal melanoma: tumour biopsy was taken 12 days after first dose (93 mg/m2); the biopsy presented spindle cell (bottom) and polygonal-shaped (top) melanoma. SCLC: tumour biopsy was taken 3 days after MRX34 dosing (93 mg/m2). HCC: tumour biopsy was taken 3 days after MRX34 dosing (70 mg/m2). GIST: tumour biopsy was taken 4 days after MRX34 dosing (93 mg/m2).
Fig. 3. Responses in patients treated with…
Fig. 3. Responses in patients treated with MRX34.
a Confirmed PR in a 32-year-old male with N-ras mutated, KIT/BRAF wild, PD-L1+ acral melanoma that was initially treated by thumb amputation. Following progression with multiple metastases, the patient was treated unsuccessfully with adoptive T-cell therapy with high-dose IL-2, ipilimumab, pembrolizumab, and CVT chemotherapy. MRX34 treatment was initiated and index lesion size reductions of 39 and 54% were observed after cycles 4 and 6, respectively. MRX34 was discontinued at the patient’s request after completion of cycle 7, after which the PR lasted an additional 7 months with no other treatment for a total duration of response of 65+ weeks. b Confirmed PR in a 56-year-old male with clear cell renal carcinoma who had been refractory to sunitinib, temsirolimus, and bevacizumab. The patient received three cycles of MRX34. Due to rising liver enzymes, treatment was discontinued at that time, and a liver biopsy showed immune hepatitis, but no tumour. PR was noted 3 months after MRX34 discontinuation and confirmed at 4.5 months. The response lasted 54 weeks.

References

    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297.
    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233.
    1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat. Rev. Cancer. 2006;6:857–866.
    1. Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat. Rev. Cancer. 2006;6:259–269.
    1. Kasinski AL, Slack FJ. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy. Nat. Rev. Cancer. 2011;11:849–864.
    1. Jansson MD, Lund AH. MicroRNA and Cancer. Mol. Oncol. 2012;6:590–610.
    1. Bader AG. miR-34–a microRNA replacement therapy is headed to the clinic. Front Genet. 2012;3:120.
    1. Cortez MA, Valdecanas D, Niknam S, et al. In vivo delivery of miR-34a sensitizes lung tumors to radiation through RAD51 regulation. Mol. Ther.—Nucleic Acids. 2015;4:e270.
    1. Bader AG, Brown D, Winkler M. The promise of microRNA replacement therapy. Cancer Res. 2010;70:7027–7030.
    1. Trang P, Wiggins JF, Daige DL, et al. Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol. Ther. 2011;19:1116–1122.
    1. Bader AG, Brown D, Stoudemire J, et al. Developing therapeutic microRNAs for cancer. Gene Ther. 2011;18:1121–1126.
    1. Daige CL, Wiggins JF, Priddy L, et al. Systemic delivery of a miR-34a mimic as a potential therapeutic for liver cancer. Mol. Cancer Ther. 2014;13:2352–2360.
    1. Kelnar K, Peltier HJ, Leatherbury N, et al. Quantification of therapeutic miRNA mimics in whole blood from non-human primates. Anal. Chem. 2014;86:1534–1542.
    1. Beg MS, Brenner AJ, Sachdev J, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest N. Drugs. 2017;35:180–188.
    1. He L, He X, Lim LP, et al. A microRNA component of the p53 tumor suppressor network. Nature. 2007;447:1130–1134.
    1. Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 2010;17:193–199.
    1. Zhao J, Lammers P, Torrance CJ, et al. TP53-independent function of miR-34a via HDAC1 and p21(CIP1/WAF1) Mol. Ther. 2013;21:678–686.
    1. Wang J, Dan G, Zhao J, et al. The predictive effect of overexpressed miR-34a on good survival of cancer patients: a systematic review and meta-analysis. Onco Targets Ther. 2015;8:2709–2719.
    1. Shin J, Danli X, Zhong XP. MicroRNA-34a enhances T cell activation by targeting diacylglycerol Kinase ζ. PLoS ONE. 2013;8:e77983.
    1. Cortez MA, Ivan C, Valdecanas D, et al. PDL1 regulation by p53 via miR-34. J. Natl Cancer Inst. 2016;108:djv303.
    1. Wang X, Li J, Dong K, et al. Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cell Signal. 2015;27:443–452.
    1. Ji Q, Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE. 2009;4:e6816.
    1. Li N, Fu H, Tie Y, et al. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett. 2009;275:44–53.
    1. Liu C, Kelnar K, Liu B, et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 2011;17:211–215.
    1. Di Martino MT, Leone E, Amodio N, et al. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: in vitro and in vivo evidence. Clin. Cancer Res. 2012;18:6260–6270.
    1. Zhao, J., Kelnar, K. & Bader, A. G. In-depth analysis shows synergy between erlotinib and miR-34a. PLoS ONE.9, e89105 (2014).
    1. Wiggins JF, Ruffino L, Kelnar K, et al. Development of a lung cancer therapeutic based on the tumor suppressor microRNA-34. Cancer Res. 2010;70:5923–5930.
    1. Craig VJ, Tzankov A, Flori M, et al. Systemic microRNA-34a delivery induces apoptosis and abrogates growth of diffuse large B-cell lymphoma in vivo. Leukemia. 2012;26:2421–2424.
    1. Tolcher AW, Rodrigueza WV, Rasco DW, et al. A phase 1 study of the BCL2-targeted deoxyribonucleic acid inhibitor (DNAi) PNT2258 in patients with advanced solid tumors. Cancer Chemother. Pharm. 2014;73:363–371.
    1. Kelnar, K. & Bader A. B. A qRT-PCR method for determining the biodistribution profile of a miR-34a mimic. in Gene Therapy of Solid Cancers: Methods and Protocols, Methods in Molecular Biology (eds Walther, W. & Stein, U.) Ch 8, Vol. 1317, 125–133, Humana Press, New York City (2015).
    1. Bartonicek N, Enright AJ. SylArray: a web server for automated detection of miRNA effects from expression data. Bioinformatics. 2010;26:2900–2901.
    1. van Dongen S, Abreu-Goodger C, Enright AJ. Detecting microRNA binding and siRNA off-target effects from expression data. Nat. Methods. 2008;5:1023–1025.
    1. Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE, et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr. Biol. 2007;17:1298–1307.
    1. Cole KA, Attiyeh EF, Mosse YP, Laquaglia MJ, Diskin SJ, Brodeur GM, et al. A functional screen identifies miR-34a as a candidate neuroblastoma tumor suppressor gene. Mol. Cancer Res. 2008;6:735–742.
    1. Ji Q, Hao X, Meng Y, Zhang M, Desano J, Fan D, et al. Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer. 2008;8:266.
    1. Lovis P, Roggli E, Laybutt DR, Gattesco S, Yang JY, Widmann C, et al. Alterations in microRNA expression contribute to fatty acid-induced pancreatic beta-cell dysfunction. Diabetes. 2008;57:2728–2736.
    1. Daige CL, Wiggins JF, Priddy L, Nelligan-Davis T, Zhao J, Brown D. Systemic delivery of a miR34a mimic as a potential therapeutic for liver cancer. Mol. Cancer Ther. 2014;13:2352–2360.
    1. Kim NH, Kim HS, Kim NG, Lee I, Choi HS, Li XY, et al. p53 and microRNA-34 are suppressors of canonical Wnt signaling. Sci. Signal. 2011;4:ra71.
    1. Rao DS, O’Connell RM, Chaudhuri AA, Garcia-Flores Y, Geiger TL, Baltimore D. MicroRNA-34a perturbs B lymphocyte development by repressing the forkhead box transcription factor Foxp1. Immunity. 2010;33:48–59.
    1. Craig VJ, Cogliatti SB, Imig J, Renner C, Neuenschwander S, Rehrauer H, et al. Myc-mediated repression of microRNA-34a promotes high-grade transformation of B-cell lymphoma by dysregulation of FoxP1. Blood. 2011;117:6227–6236.
    1. Zhao J, Lammers P, Torrance CJ, Bader AG. TP53-independent function of miR-34a via HDAC1 and p21CIP1/WAF1. Mol. Ther. 2013;21:1678–1686.
    1. Robbins M, Judge A, Ambegia E, et al. Misinterpreting the therapeutic effects of small interfering RNA caused by immune stimulation. Hum. Gene Ther. 2008;19:991–999.
    1. Chattopadhyay S, Sen GC. dsRNA-activation of TLR3 and RLR signaling: gene induction-dependent and independent effects. J. Interf. Cytokine Res. 2014;34:427–436.
    1. Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162:974–986.
    1. Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 2009;15:7412–7420.
    1. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J. Clin. Oncol. 2015;33:1974–1982.
    1. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015;372:2521–2532.
    1. Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 2018;378:1277–1290.
    1. El-Khoueiry AB, Sangro B, Yau T, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. 2017;389:2492–2502.

Source: PubMed

3
Předplatit