Phase Ib dose-escalation study of the hypoxia-modifier Myo-inositol trispyrophosphate in patients with hepatopancreatobiliary tumors

Marcel A Schneider, Michael Linecker, Ralph Fritsch, Urs J Muehlematter, Daniel Stocker, Bernhard Pestalozzi, Panagiotis Samaras, Alexander Jetter, Philipp Kron, Henrik Petrowsky, Claude Nicolau, Jean-Marie Lehn, Bostjan Humar, Rolf Graf, Pierre-Alain Clavien, Perparim Limani, Marcel A Schneider, Michael Linecker, Ralph Fritsch, Urs J Muehlematter, Daniel Stocker, Bernhard Pestalozzi, Panagiotis Samaras, Alexander Jetter, Philipp Kron, Henrik Petrowsky, Claude Nicolau, Jean-Marie Lehn, Bostjan Humar, Rolf Graf, Pierre-Alain Clavien, Perparim Limani

Abstract

Hypoxia is prominent in solid tumors and a recognized driver of malignancy. Thus far, targeting tumor hypoxia has remained unsuccessful. Myo-inositol trispyrophosphate (ITPP) is a re-oxygenating compound without apparent toxicity. In preclinical models, ITPP potentiates the efficacy of subsequent chemotherapy through vascular normalization. Here, we report the results of an unrandomized, open-labeled, 3 + 3 dose-escalation phase Ib study (NCT02528526) including 28 patients with advanced primary hepatopancreatobiliary malignancies and liver metastases of colorectal cancer receiving nine 8h-infusions of ITPP over three weeks across eight dose levels (1'866-14'500 mg/m2/dose), followed by standard chemotherapy. Primary objectives are assessment of the safety and tolerability and establishment of the maximum tolerated dose, while secondary objectives include assessment of pharmacokinetics, antitumor activity via radiological evaluation and assessment of circulatory tumor-specific and angiogenic markers. The maximum tolerated dose is 12,390 mg/m2, and ITPP treatment results in 32 treatment-related toxicities (mostly hypercalcemia) that require little or no intervention. 52% of patients have morphological disease stabilization under ITPP monotherapy. Following subsequent chemotherapy, 10% show partial responses while 60% have stable disease. Decreases in angiogenic markers are noted in ∼60% of patients after ITPP and tend to correlate with responses and survival after chemotherapy.

Conflict of interest statement

C.N. is co-founder, director, and member of the scientific advisory board of NormOxys ®, Inc. and J.-M.L. is co-founder and chairman of scientific advisory board of NormOxys ®, Inc, this being the company that holds the patent on ITPP. M.A.S., P.L., R.G., B.H. and P.-A.C. had access to raw data. The study drug was provided by NormOxys ®, Inc, free of charge. Neither NormOxys ®, Inc nor any other funding source were involved in study design, patient recruitment/care, data collection, analysis/interpretation, or manuscript writing. The remaining authors declare no competing interests.

Figures

Fig. 1. Study setup and pharmacokinetics.
Fig. 1. Study setup and pharmacokinetics.
a Schema of study flow with timepoints of ITPP administration and assessments. b Dose-escalation schema with single, weekly, and total doses for different cohorts. c Boxplots displaying median plasma concentrations at start of infusion (hour 0), 3 and 6 h as wells as 30 min, 1 and 2 h after end of intravenous ITPP administration (hour 8) of increasing doses in cohorts on treatment days. Upper and lower ends of boxplots represent 25th and 75th quartiles. Whiskers extend to values within 1.5 * IQR from the boxplot, with data beyond plotted separately as outliers (n = 3–4 patients per cohort with 1 measurement performed in technical duplicates per timepoint for each treatment day (normally 9) per patient).
Fig. 2. Radiological responses post ITPP and…
Fig. 2. Radiological responses post ITPP and chemotherapy.
Displayed as waterfall plots of percental changes of either diameter in millimeters for RECIST1.1 criteria or SUV uptake for EORTC criteria. Morphological changes according to RECIST1.1 criteria (a) and metabolic changes according to EORTC criteria (b) in target lesions after ITPP monotherapy. Morphological changes according to RECIST1.1 criteria (c) and metabolic changes according to EORTC criteria (d) in target lesions after subsequent chemotherapy. *Indicates the appearance of new (FDG avid) lesions.
Fig. 3. Changes in serum angiogenic markers…
Fig. 3. Changes in serum angiogenic markers post ITPP and correlation with chemotherapy responses and survival.
Changes in circulating levels of VEGFA (a), ANG1 (b), ANG2 (c), EGF (d), and PECAM1/CD31 (e). Percental changes of pre- versus post-ITPP monotherapy markers levels are depicted (from left to right) by patient as waterfall plots. Association of morphological and metabolic response post chemotherapy with changes in serum angiogenesis markers under ITPP treatment are shown as boxplots displaying median values with the upper and lower ends representing the 25th and 75th quartiles, respectively. Whiskers extend to values within 1.5 * IQR from the boxplot, with all individual data points shown overlaid and colored according to tumor type. Survival stratified by decreased or increased marker levels displayed as Kaplan–Meier curves. n = 27 individual patients, marker measurements performed as technical duplicates.
Fig. 4. Tissue-based hypoxia response markers of…
Fig. 4. Tissue-based hypoxia response markers of one exemplary patient.
a Histological images taken at 20x magnification of the sample taken before ITPP treatment (left hemi-hepatectomy) compared to the sample obtained after ITPP (microwave ablation). Note the decreased expression of hypoxia-mediated genes such as CA9 and SLC2A1 after ITPP treatment and signs of increased vessel maturity by increased EGR expression. Scale bare: 200 μm. N = 1 patient. Histological quantifications are shown as boxplots displaying mean values ± standard deviation and overlaid single measurements. Statistical differences derived from two-sided students t-test with no adjustment for multiple comparisons. **p < 0.01, ***p ≤ 0.001, NS. = No significant difference.

References

    1. Pouyssegur J, Dayan F, Mazure NM. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature. 2006;441:437–443. doi: 10.1038/nature04871.
    1. Bhandari V, et al. Molecular landmarks of tumor hypoxia across cancer types. Nat. Genet. 2019;51:308–318. doi: 10.1038/s41588-018-0318-2.
    1. De Bock K, Mazzone M, Carmeliet P. Antiangiogenic therapy, hypoxia, and metastasis: risky liaisons, or not? Nat. Rev. Clin. Oncol. 2011;8:393–404. doi: 10.1038/nrclinonc.2011.83.
    1. Noman MZ, et al. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am. J. Physiol. Cell Physiol. 2015;309:C569–C579. doi: 10.1152/ajpcell.00207.2015.
    1. Walsh JC, et al. The clinical importance of assessing tumor hypoxia: relationship of tumor hypoxia to prognosis and therapeutic opportunities. Antioxid. Redox Signal. 2014;21:1516–1554. doi: 10.1089/ars.2013.5378.
    1. Kerbel RS. Tumor angiogenesis. N. Engl. J. Med. 2008;358:2039–2049. doi: 10.1056/NEJMra0706596.
    1. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer. 2011;11:393–410. doi: 10.1038/nrc3064.
    1. Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to hypoxic stress. Mol. cell. 2010;40:294–309. doi: 10.1016/j.molcel.2010.09.022.
    1. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N. Engl. J. Med. 2011;364:656–665. doi: 10.1056/NEJMra0910283.
    1. Al Tameemi W, Dale TP, Al-Jumaily RMK, Forsyth NR. Hypoxia-modified cancer cell metabolism. Front Cell Dev. Biol. 2019;7:4. doi: 10.3389/fcell.2019.00004.
    1. Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Science. 2016;352:175–180. doi: 10.1126/science.aaf4405.
    1. Rankin EB, Nam JM, Giaccia AJ. Hypoxia: signaling the metastatic cascade. Trends Cancer. 2016;2:295–304. doi: 10.1016/j.trecan.2016.05.006.
    1. Noman MZ, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014;211:781–790. doi: 10.1084/jem.20131916.
    1. Chouaib S, Noman MZ, Kosmatopoulos K, Curran MA. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene. 2017;36:439–445. doi: 10.1038/onc.2016.225.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Ye Y, et al. Characterization of hypoxia-associated molecular features to aid hypoxia-targeted therapy. Nat. Metab. 2019;1:431–444. doi: 10.1038/s42255-019-0045-8.
    1. Hurwitz H, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691.
    1. Paez-Ribes M, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15:220–231. doi: 10.1016/j.ccr.2009.01.027.
    1. Welsh S, Williams R, Kirkpatrick L, Paine-Murrieta G, Powis G. Antitumor activity and pharmacodynamic properties of PX-478, an inhibitor of hypoxia-inducible factor-1alpha. Mol. Cancer Therapeutics. 2004;3:233–244.
    1. Zhao T, et al. Inhibition of HIF-1α by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget. 2015;6:2250–2262. doi: 10.18632/oncotarget.2948.
    1. Schwartz DL, et al. Radiosensitization and stromal imaging response correlates for the HIF-1 inhibitor PX-478 given with or without chemotherapy in pancreatic cancer. Mol. Cancer Therapeutics. 2010;9:2057–2067. doi: 10.1158/1535-7163.MCT-09-0768.
    1. Tibes R, et al. Results from a phase I, dose-escalation study of PX-478, an orally available inhibitor of HIF-1α. J. Clin. Oncol. 2010;28:3076–3076. doi: 10.1200/jco.2010.28.15_suppl.3076.
    1. Borad MJ, et al. Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. J. Clin. Oncol. 2015;33:1475–1481. doi: 10.1200/JCO.2014.55.7504.
    1. Kishimoto S. et al. Hypoxia-activated prodrug evofosfamide treatment in pancreatic ductal adenocarcinoma xenografts alters the tumor redox status to potentiate radiotherapy. Antioxid. Redox Signal.10.1089/ars.2020.8131 (2020). Accession Number: 32787454.
    1. Sun JD, et al. Efficacy and safety of the hypoxia-activated prodrug TH-302 in combination with gemcitabine and nab-paclitaxel in human tumor xenograft models of pancreatic cancer. Cancer Biol. Ther. 2015;16:438–449. doi: 10.1080/15384047.2014.1003005.
    1. Yoon, J., Kang, S. Y., Lee, K. H., Cheon, G. J., Oh, D. Y. Targeting hypoxia using evofosfamide and companion hypoxia imaging of FMISO-PET in advanced biliary tract cancer. Cancer Res. Treatment53, 471–479 (2021).
    1. Duran R, et al. Preclinical benefit of hypoxia-activated intra-arterial therapy with evofosfamide in liver cancer. Clin. Cancer Res. 2017;23:536–548. doi: 10.1158/1078-0432.CCR-16-0725.
    1. Haynes J, et al. Administration of hypoxia-activated prodrug evofosfamide after conventional adjuvant therapy enhances therapeutic outcome and targets cancer-initiating cells in preclinical models of colorectal cancer. Clin. Cancer Res. 2018;24:2116–2127. doi: 10.1158/1078-0432.CCR-17-1715.
    1. Chawla SP, et al. Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. J. Clin. Oncol. 2014;32:3299–3306. doi: 10.1200/JCO.2013.54.3660.
    1. Ganjoo KN, et al. A phase I study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology. 2011;80:50–56. doi: 10.1159/000327739.
    1. Sun JD, et al. Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer. Clin. Cancer Res. 2012;18:758–770. doi: 10.1158/1078-0432.CCR-11-1980.
    1. Tap WD, et al. Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2017;18:1089–1103. doi: 10.1016/S1470-2045(17)30381-9.
    1. Welsh L, et al. Blood transfusion during radical chemo-radiotherapy does not reduce tumour hypoxia in squamous cell cancer of the head and neck. Br. J. Cancer. 2017;116:28–35. doi: 10.1038/bjc.2016.386.
    1. Davidson A, et al. A phase III randomized trial of adding topical nitroglycerin to first-line chemotherapy for advanced nonsmall-cell lung cancer: the Australasian lung cancer trials group NITRO trial. Ann. Oncol. 2015;26:2280–2286. doi: 10.1093/annonc/mdv373.
    1. Gupta N, et al. Carbogen and nicotinamide increase blood flow and 5-fluorouracil delivery but not 5-fluorouracil retention in colorectal cancer metastases in patients. Clin. Cancer Res. 2006;12:3115–3123. doi: 10.1158/1078-0432.CCR-05-0513.
    1. Heys SD, et al. A pilot study with long term follow up of hyperbaric oxygen pretreatment in patients with locally advanced breast cancer undergoing neo-adjuvant chemotherapy. Undersea Hyperbaric Med. 2006;33:33–43.
    1. Duarte CD, Greferath R, Nicolau C, Lehn JM. myo-Inositol trispyrophosphate: a novel allosteric effector of hemoglobin with high permeation selectivity across the red blood cell plasma membrane. Chembiochem. 2010;11:2543–2548. doi: 10.1002/cbic.201000499.
    1. Kieda C, et al. Stable tumor vessel normalization with pO2 increase and endothelial PTEN activation by inositol trispyrophosphate brings novel tumor treatment. J. Mol. Med. 2013;91:883–899. doi: 10.1007/s00109-013-0992-6.
    1. Raykov Z, et al. Myo-inositol trispyrophosphate-mediated hypoxia reversion controls pancreatic cancer in rodents and enhances gemcitabine efficacy. Int. J. Cancer. 2014;134:2572–2582. doi: 10.1002/ijc.28597.
    1. Limani P, et al. Antihypoxic potentiation of standard therapy for experimental colorectal liver metastasis through myo-inositol trispyrophosphate. Clin. Cancer Res. 2016;22:5887–5897. doi: 10.1158/1078-0432.CCR-15-3112.
    1. Limani P, et al. The allosteric hemoglobin effector ITPP inhibits metastatic colon cancer in mice. Ann. Surg. 2017;266:746–753. doi: 10.1097/SLA.0000000000002431.
    1. Derbal-Wolfrom L, et al. Increasing the oxygen load by treatment with myo-inositol trispyrophosphate reduces growth of colon cancer and modulates the intestine homeobox gene Cdx2. Oncogene. 2013;32:4313–4318. doi: 10.1038/onc.2012.445.
    1. Aprahamian M, et al. Myo-InositolTrisPyroPhosphate treatment leads to HIF-1α suppression and eradication of early hepatoma tumors in rats. Chembiochem. 2011;12:777–783. doi: 10.1002/cbic.201000619.
    1. Bao B, et al. In vivo imaging and quantification of carbonic anhydrase IX expression as an endogenous biomarker of tumor hypoxia. PLoS ONE. 2012;7:e50860. doi: 10.1371/journal.pone.0050860.
    1. Tafreshi N. K., Lloyd M. C., Bui M. M., Gillies R. J., Morse D. L. Carbonic anhydrase IX as an imaging and therapeutic target for tumors and metastases. In: Carbonic Anhydrase: Mechanism, Regulation, Links to Disease, and Industrial Applications (eds. Frost, S. C. and McKenna, R.) (Springer Netherlands, 2014).
    1. Yu M, et al. The prognostic value of GLUT1 in cancers: a systematic review and meta-analysis. Oncotarget. 2017;8:43356–43367. doi: 10.18632/oncotarget.17445.
    1. Shah AV, Birdsey GM, Randi AM. Regulation of endothelial homeostasis, vascular development and angiogenesis by the transcription factor ERG. Vasc. Pharmacol. 2016;86:3–13. doi: 10.1016/j.vph.2016.05.003.
    1. Biolo A, et al. Enhanced exercise capacity in mice with severe heart failure treated with an allosteric effector of hemoglobin, myo-inositol trispyrophosphate. Proc. Natl Acad. Sci. USA. 2009;106:1926–1929. doi: 10.1073/pnas.0812381106.
    1. Chung AS, Lee J, Ferrara N. Targeting the tumour vasculature: insights from physiological angiogenesis. Nat. Rev. Cancer. 2010;10:505–514. doi: 10.1038/nrc2868.
    1. Fagiani E, Christofori G. Angiopoietins in angiogenesis. Cancer Lett. 2013;328:18–26. doi: 10.1016/j.canlet.2012.08.018.
    1. Khong TL, et al. Identification of the angiogenic gene signature induced by EGF and hypoxia in colorectal cancer. BMC Cancer. 2013;13:518. doi: 10.1186/1471-2407-13-518.
    1. Zafar N, et al. Circulating angiogenic stem cells in type 2 diabetes are associated with glycemic control and endothelial dysfunction. PLoS ONE. 2018;13:e0205851. doi: 10.1371/journal.pone.0205851.
    1. Liu Z., et al. CD31-labeled circulating endothelial cells as predictor in anlotinib-treated non-small-cell lung cancer: analysis on ALTER-0303 study. Cancer Med. 10.1002/cam4.1584 (2018). Accession Number: 29856135.
    1. Dewhirst MW. Intermittent hypoxia furthers the rationale for hypoxia-inducible factor-1 targeting. Cancer Res. 2007;67:854–855. doi: 10.1158/0008-5472.CAN-06-4744.
    1. Limani P, et al. Development of OXY111A, a novel hypoxia-modifier as a potential antitumor agent in patients with hepato-pancreato-biliary neoplasms - Protocol of a first Ib/IIa clinical trial. BMC Cancer. 2016;16:812. doi: 10.1186/s12885-016-2855-3.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Young H, et al. Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group. Eur. J. Cancer. 1999;35:1773–1782. doi: 10.1016/S0959-8049(99)00229-4.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods. 2012;9:671–675. doi: 10.1038/nmeth.2089.

Source: PubMed

3
Předplatit