A randomized phase II study of SM-88 plus methoxsalen, phenytoin, and sirolimus in patients with metastatic pancreatic cancer treated in the second line and beyond

Marcus S Noel, Semmie Kim, Marion L Hartley, Steve Wong, Vincent J Picozzi, Harry Staszewski, Dae Won Kim, Jan M Van Tornout, Philip Agop Philip, Vincent Chung, Allyson J Ocean, Andrea Wang-Gillam, Marcus S Noel, Semmie Kim, Marion L Hartley, Steve Wong, Vincent J Picozzi, Harry Staszewski, Dae Won Kim, Jan M Van Tornout, Philip Agop Philip, Vincent Chung, Allyson J Ocean, Andrea Wang-Gillam

Abstract

Background: This trial explores SM-88 used with methoxsalen, phenytoin, and sirolimus (MPS) in pretreated metastatic pancreatic ductal adenocarcinoma (mPDAC) METHODS: Forty-nine patients were randomized to daily 460 or 920 mg oral SM-88 with MPS (SM-88 Regimen). The primary endpoint was objective response rate (RECIST 1.1).

Results: Thirty-seven patients completed ≥ one cycle of SM-88 Regimen (response evaluable population). Disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) did not differ significantly between dose levels. Stable disease was achieved in 9/37 patients (DCR, 24.3%); there were no complete or partial responses. Quality-of-life (QOL) was maintained and trended in favor of 920 mg. SM-88 Regimen was well tolerated; a single patient (1/49) had related grade 3 and 4 adverse events, which later resolved. In the intention-to-treat population of 49 patients, the median overall survival (mOS) was 3.4 months (95% CI: 2.7-4.9 months). Those treated in the second line had an mOS of 8.1 months and a median PFS of 3.8 months. Survival was higher for patients with stable versus progressive disease (any line; mOS: 10.6 months vs. 3.9 months; p = 0.01).

Conclusions: SM-88 Regimen has a favorable safety profile with encouraging QOL effects, disease control, and survival trends. This regimen should be explored in the second-line treatment of patients with mPDAC.

Clinicaltrials: gov Identifier: NCT03512756.

Conflict of interest statement

Dr. Marcus Smith Noel declares that he has held a Consulting or Advisory Role at Celgene, Taiho Pharmaceutical, and Ipsen. He is also a member of the Speakers Bureau for Celgene, Taiho Pharmaceutical, and Daiichi Sankyo/Astra Zeneca.

Semmie Kim declares Employment, Stock, and other Ownership Interests from TYME Inc.

Marion L. Hartley, PhD declares no potential competing interests.

Dr. Steve Wong declares no potential competing interests.

Dr. Vincent Picozzi declares that he has held a Consulting or Advisory Role at TriSalus Life Sciences. He also has Stock and other Ownership Interests in Amgen, Johnson and Johnson, McKesson, Thermo Fisher Scientific, and Cigna. He has received Research Funding from FibroGen, Celgene, Ipsen, NovoCure, Merk, TYME Inc., Abbvie, and NGM Biopharmaceuticals.

Dr. Harry Staszewski declare no potential competing interests.

Dr. Dae Won Kim has received Research Funding from Bold Therapeutics.

Dr. Jan Van Tornout declares employment at TYME Inc. Stock and other Ownership Interests from Bristol‐Myers Squibb, Novartis, Sanofi, TYME Inc., Lilly, and Merck. Has received an Honoraria from TYME Inc. Has held a Consulting or Advisory Role at TYME Inc.

Dr. Philip Agop Philip has held a Consulting or Advisory Role at Celgene, Ipsen, Merck, TriSalus, Daiichi, Syncore, and Taiho. He has been on the Speakers' Bureau for Celgene, Bayer, Ipsen, Novartis, and Incyte. Has received reimbursement for travel, accommodation, and expenses from Rafael Pharmaceuticals, Celgene, and Abbvie. He has received an Honoraria from Celgene, Bayer, Ipsen, Merck, AstraZeneca, TriSalus Life Sciences, Blueprint Medicines, Syncore, and Array BioPharma. He has received Research Funding from Bayer, Incyte, Karyopharm Therapeutics, Merck, Taiho Pharmaceutical, Momenta Pharmaceuticals, Novartis, Plexxikon, Immunomedics, Regeneron, Genentech, TYME Inc., Caris Life Sciences, ASLAN Pharmaceuticals, QED Therapeutics, Halozyme, Boston Biomedical, Advanced Accelerator Applications, Lilly, Taiho Pharmaceuticals, and Merus.

Dr. Vincent Chung has held a Consulting or Advisory Role at Ispen, Gristone Oncology, Westwood Bioscience, and Apeiron Biologics. He has been on the Speakers' Bureau for Ipsen and Celgene. He has received Research Funding from Roche and Merck.

Dr. Allyson J. Ocean has held a Consulting or Advisory Role at TYME Inc., and Celgene. She has been on the Speakers' Bureau at Daiichi Sankyo. Has received reimbursement for travel, accommodation, and expenses from Daiichi Sankyo.

Dr. Andrea Wang‐Gillam declares employment at Jocobio. She has held a Consulting or Advisory Role at AstraZeneca, Merck, Bayer, and Inxmed. Stock and other Ownership Interests at Jacobio. She has received Research Funding from AstraZeneca, Pfizer, Lilly, Verastem, Merck, BioMed Valley Discoveries, Roche, Bristol‐Myers Squibb, Xcovery, Boston Biomedical, Hutchinson MediPharma, Rafael Pharmaceuticals, Gossamer Bio.

© 2022 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Patient overall quality‐of‐life scores on treatment, as measured by the EORTC QLQ‐C30 Question 30, and weight by the dose of SM‐88 Regimen. Box and whisker plots showing median, interquartile range, and minimum and maximum values of (A) EORTC QLQ‐C30 Question 30, “How would you rate your overall QOL during the past week?” from pre‐treatment to Cycle 4 on treatment; and (B) weight in pounds pre‐ and post‐treatment by dose group. In (A), scoring ranges from 1 to 7, where 1 = very poor and 7 = excellent. EORTC QLQ‐C30, European Organization for Research and Treatment of Cancer Quality‐of‐Life Questionnaire; QOL, quality‐of‐life
FIGURE 2
FIGURE 2
Overall survival (OS) of evaluable patients receiving SM‐88 Regimen (n = 37). Simple Kaplan– Meier curve of OS in months for (A) all 37 patients (median 3.9 months; 95% CI: 3.0–5.7), (B) the 20 patients receiving 460 mg/day compared to the 17 patients receiving 920 mg/day (4.9 months [95% CI: 2.2–8.1] vs. 3.6 months [95% CI: 2.6–5.7]; p = 0.79) and (C) patients receiving SM‐88 Regimen after 1 prior line (n = 5), 2 prior lines (n = 18), and 3 or more prior lines (n = 14) of therapy: 8.1 months (95% CI: 3.0 ‐ n/a [not reached]) vs. 3.7 months (95% CI: 2.7–6.6) versus 3.0 months (95% CI: 1.9–5.4); log‐rank p = 0.08
FIGURE 3
FIGURE 3
OS of evaluable patients by RECIST best response (SD or PD). Kaplan–Meier OS curve for evaluable patients receiving SM‐88 Regimen who achieved stable disease (solid line; mOS = 10.6 months [95% CI: 2.6–30.5]) versus those who had disease progression (hashed line; mOS = 3.9 months [95% CI: 3.0–5.7]). Log‐rank p = 0.01; HR = 0.29 (95% CI: 0.10–0.79). mOS, median overall survival; OS, overall survival; RECIST, response evaluation criteria in solid tumors; SD, stable disease
FIGURE 4
FIGURE 4
PFS curves for all evaluable patients (n = 37). Kaplan–Meier curve of PFS in months for (A) all 37 evaluable patients (median 1.9 months [95% CI: 1.7‐2.0]), (B) the 20 patients receiving 460 mg/day (1.9 months [95% CI: 1.6–3.0]) compared to the 17 patients receiving 920 mg/day (1.8 months [95% CI: 1.0–2.6]; log‐rank p = 0.25), and (C) patients receiving SM‐88 Regimen after 1 prior line (solid line; n = 5), 2 prior lines (black hashed line; n = 18), and 3 or more prior lines (black dotted line; n = 14) of therapy (3.8 months [95% CI: 0.9 – not reached], 1.8 months [95% CI: 1.5–2.0], and 1.9 months [95% CI: 1.4–2.6]; log‐rank p = 0.44). *A single patient who had received 2 lines of prior treatment showed a prolonged PFS response, with RECIST‐confirmed stable disease at 15 weeks post‐treatment initiation. PFS, progression‐free survival; RECIST, response evaluation criteria in solid tumors

References

    1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 Countries. CA Cancer J Clin. 2021;71:209‐249.
    1. Howlader N, Noone AM, Krapcho M, et al., eds. SEER Cancer Statistics Review, 1975–2016. National Cancer Institute. Bethesda, MD, based on November 2018 SEER data submission, posted to the SEER Web site, April 2019 . Accessed November 9, 2021.
    1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7‐33.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7‐30.
    1. Rahib L, Wehner MR, Matrisian LM, Nead KT. Estimated projection of US cancer incidence and death to 2040. JAMA Netw Open. 2021;4:e214708.
    1. Krantz BA, Yu KH, O'Reilly EM. Pancreas adenocarcinoma: novel therapeutics. Chin Clin Oncol. 2017;6:30.
    1. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817‐1825.
    1. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab‐paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691‐1703.
    1. Wang‐Gillam A, Li CP, Bodoky G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine‐based therapy (NAPOLI‐1): a global, randomised, open‐label, phase 3 trial. Lancet. 2016;387:545‐557.
    1. Moriya K, Nakagawa K, Santa T, et al. Oxidative stress in the absence of inflammation in a mouse model for hepatitis C virus‐associated hepatocarcinogenesis. Cancer Res. 2001;61:4365‐4370.
    1. Wiseman H, Halliwell B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem J. 1996;313(Pt 1):17‐29.
    1. Akladios FN, Andrew SD, Parkinson CJ. Selective induction of oxidative stress in cancer cells via synergistic combinations of agents targeting redox homeostasis. Bioorg Med Chem. 2015;23:3097‐3104.
    1. Epstein T, Xu L, Gillies RJ, Gatenby RA. Separation of metabolic supply and demand: aerobic glycolysis as a normal physiological response to fluctuating energetic demands in the membrane. Cancer Metab. 2014;2:7.
    1. Kryston TB, Georgiev AB, Pissis P, Georgakilas AG. Role of oxidative stress and DNA damage in human carcinogenesis. Mutat Res. 2011;711:193‐201.
    1. Kaira K, Oriuchi N, Otani Y, et al. Fluorine‐18‐alpha‐methyltyrosine positron emission tomography for diagnosis and staging of lung cancer: a clinicopathologic study. Clin Cancer Res. 2007;13:6369‐6378.
    1. Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF‐4E BP1 through a common effector mechanism. J Biol Chem. 1998;273:14484‐14494.
    1. Lynch CJ, Fox HL, Vary TC, Jefferson LS, Kimball SR. Regulation of amino acid‐sensitive TOR signaling by leucine analogues in adipocytes. J Cell Biochem. 2000;77:234‐251.
    1. Wiriyasermkul P, Nagamori S, Tominaga H, et al. Transport of 3‐fluoro‐L‐alpha‐methyl‐tyrosine by tumor‐upregulated L‐type amino acid transporter 1: a cause of the tumor uptake in PET. J Nucl Med. 2012;53:1253‐1261.
    1. May ME, Buse MG. Effects of branched‐chain amino acids on protein turnover. Diabetes Metab Rev. 1989;5:227‐245.
    1. Mortimore GE, Pösö AR, Kadowaki M, Wert JJ Jr. Multiphasic control of hepatic protein degradation by regulatory amino acids. General features and hormonal modulation. J Biol Chem. 1987;262:16322‐16327.
    1. Shigemitsu K, Tsujishita Y, Hara K, Nanahoshi M, Avruch J, Yonezawa K. Regulation of translational effectors by amino acid and mammalian target of rapamycin signaling pathways. Possible involvement of autophagy in cultured hepatoma cells. J Biol Chem. 1999;274:1058‐1065.
    1. Wei L, Tominaga H, Ohgaki R, et al. Specific transport of 3‐fluoro‐l‐α‐methyl‐tyrosine by LAT1 explains its specificity to malignant tumors in imaging. Cancer Sci. 2016;107:347‐352.
    1. Stega J, Noel MS, Vandell AG, Stega D, del Priore G, Hoffman S. A first‐in‐human study of the novel metabolism‐based anti‐cancer agent SM‐88 in subjects with advanced metastatic cancer. Invest New Drugs. 2019;38:392‐401.
    1. Hoffman S, Stega J, Zawisny P, et al. SM‐88 therapy in patients with advanced or metastatic pancreatic cancer. J Clin Oncol. 2018;36(4_supp):457–457.
    1. Chua VS, Chawla SP, Zheng K, Kim T, del Priore G, Kim S. Phase II study of SM‐88 in Ewing's and other sarcomas. J Clin Oncol. 2021;39:e23505–e23505.
    1. Gartrell BA, Roach M III, Retter A, Sokol GH, del Priore G, Scher HI. Phase II trial of SM‐88, a cancer metabolism based therapy, in non‐metastatic biochemical recurrent prostate cancer. Invest New Drugs. 2021;39:499‐508.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228‐247.
    1. National Cancer Institute, National Institutes of Health, US Department of Health and Human Services . Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. Published May 28, 2009; Revised version 4.03 June 14, 2010. Accessed May 20, 2013.
    1. American Cancer Society . Cancer Facts & Figures 2021. American Cancer Society; 2021. Accessed November 9, 2021.
    1. Zaniboni A, Aitini E, Barni S, et al. FOLFIRI as second‐line chemotherapy for advanced pancreatic cancer: a GISCAD multicenter phase II study. Cancer Chemother Pharmacol. 2012;69:1641‐1645.
    1. Xiong HQ, Varadhachary GR, Blais JC, Hess KR, Abbruzzese JL, Wolff RA. Phase 2 trial of oxaliplatin plus capecitabine (XELOX) as second‐line therapy for patients with advanced pancreatic cancer. Cancer. 2008;113:2046‐2052.
    1. Gill S, Ko YJ, Cripps C, et al. PANCREOX: a randomized phase III study of fluorouracil/leucovorin with or without oxaliplatin for second‐line advanced pancreatic cancer in patients who have received gemcitabine‐based chemotherapy. J Clin Oncol. 2016;34:3914‐3920.
    1. Yoo C, Hwang JY, Kim JE, et al. A randomised phase II study of modified FOLFIRI.3 vs modified FOLFOX as second‐line therapy in patients with gemcitabine‐refractory advanced pancreatic cancer. Br J Cancer. 2009;101:1658‐1663.
    1. Wang‐Gillam A, Hubner RA, Siveke JT, et al. NAPOLI‐1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: final overall survival analysis and characteristics of long‐term survivors. Eur J Cancer. 2019;108:78‐87.
    1. Katopodis O, Polyzos A, Kentepozidis N, et al. Second‐line chemotherapy with capecitabine (Xeloda) and docetaxel (Taxotere) in previously treated, unresectable adenocarcinoma of pancreas: the final results of a phase II trial. Cancer Chemother Pharmacol. 2011;67:361‐368.
    1. Mita N, Iwashita T, Uemura S, et al. Second‐line gemcitabine plus nab‐paclitaxel for patients with unresectable advanced pancreatic cancer after first‐line FOLFIRINOX failure. J Clin Med. 2019;8:761.
    1. Hammel P, Fabienne P, Mineur L, et al. Erythrocyte‐encapsulated asparaginase (eryaspase) combined with chemotherapy in second‐line treatment of advanced pancreatic cancer: an open‐label, randomized Phase IIb trial. Eur J Cancer. 2020;124:91‐101.
    1. Erytech . Accessed February 10, 2022. . 10/25/21.
    1. Renouf DJ, Tang PA, Hedley D, et al. A phase II study of erlotinib in gemcitabine refractory advanced pancreatic cancer. Eur J Cancer. 2014;50:1909‐1915.
    1. Brell JM, Matin K, Evans T, et al. Phase II study of docetaxel and gefitinib as second‐line therapy in gemcitabine pretreated patients with advanced pancreatic cancer. Oncology. 2009;76:270‐274.
    1. Bockorny B, Semenisty V, Macarulla T, et al. BL‐8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26:878‐885.
    1. O'Reilly EM, Oh DY, Dhani N, et al. Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2019;5:1431‐1438.
    1. Le DT, Picozzi VJ, Ko AH, et al. Results from a phase IIb, randomized, multicenter study of GVAX pancreas and CRS‐207 compared with chemotherapy in adults with previously treated metastatic pancreatic adenocarcinoma (ECLIPSE study). Clin Cancer Res. 2019;25:5493‐5502.
    1. Cardin DB, Thota R, Goff LW, et al. A phase II study of ganetespib as second‐line or third‐line therapy for metastatic pancreatic cancer. Am J Clin Oncol. 2018;41:772‐776.
    1. Weinberg BA, Wang H, Witkiewicz AK, et al. A phase I study of ribociclib plus everolimus in patients with metastatic pancreatic adenocarcinoma refractory to chemotherapy. J Pancreat Cancer. 2020;6:45‐54.
    1. Picozzi VJ, Ramanathan RK, Lowery MA, et al. 90Y‐clivatuzumab tetraxetan with or without low‐dose gemcitabine: a phase Ib study in patients with metastatic pancreatic cancer after two or more prior therapies. Eur J Cancer. 2015;51:1857‐1864.
    1. Immunomedics . March 14, 2016. Accessed November 16, 2021.
    1. Gourgou‐Bourgade S, Bascoul‐Mollevi C, Desseigne F, et al. Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31:23‐29.
    1. Prigerson HG, Bao Y, Shah MA, et al. Chemotherapy use, performance status, and quality of life at the end of life. JAMA Oncol. 2015;1:778‐784.
    1. Oettle H, Riess H, Stieler JM, et al. Second‐line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine‐refractory pancreatic cancer: outcomes from the CONKO‐003 trial. J Clin Oncol. 2014;32:2423‐2429.
    1. Hurwitz HI, Uppal N, Wagner SA, et al. Randomized, double‐blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed. J Clin Oncol. 2015;33:4039‐4047.

Source: PubMed

3
订阅