Skip to main content
Top
Published in: Investigational New Drugs 4/2019

01-08-2019 | PHASE I STUDIES

A phase I study of the safety and tolerability of VLX600, an Iron Chelator, in patients with refractory advanced solid tumors

Authors: Kabir Mody, Aaron S. Mansfield, Lalitha Vemireddy, Peter Nygren, Joachim Gulbo, Mitesh Borad

Published in: Investigational New Drugs | Issue 4/2019

Login to get access

Summary

Introduction VLX600 is a novel iron chelator designed to interfere with intracellular iron metabolism, leading to inhibition of mitochondrial respiration and bioenergetic catastrophe and resultant tumor cell death. Methods We conducted a multicenter, phase 1, dose escalation study to determine the safety and adverse event profile and the maximum tolerated dose and recommended phase 2 dose of VLX600. Other endpoints included pharmacokinetics, and preliminary evidence of anti-cancer efficacy as assessed according to RECIST 1.1 criteria. VLX600 was administered intravenously on days 1, 8, and 15 of each 28-day treatment cycle. Results Nineteen patients were enrolled, and seventeen received at least one dose of VLX600. Dose increments were reduced to 50% after dose level 3 (40 mg) due to the occurrence of a grade 3 pulmonary embolism. The study was then closed early due to slow recruitment. No maximum tolerated dose (MTD) nor RP2D had been identified at the time of study closure. Overall, the drug was well tolerated and no DLTs were observed. Fourteen patients experienced drug-related adverse events of any grade. The most frequently reported drug-related AEs were fatigue, nausea, constipation, vomiting, increased alkaline phosphatase, anemia, and decreased appetite. No formal efficacy or survival analyses were performed. No objective responses were observed, though six patients (32%) had stable disease as best response. Conclusion VLX600 was reasonably well tolerated and, together with preclinical data, there is support for further efforts to explore its activity as single agent and in combination with drugs or radiation.
Literature
1.
go back to reference Mellor HR, Callaghan R (2008) Resistance to chemotherapy in cancer: a complex and integrated cellular response. Pharmacology 81(4):275–300CrossRefPubMed Mellor HR, Callaghan R (2008) Resistance to chemotherapy in cancer: a complex and integrated cellular response. Pharmacology 81(4):275–300CrossRefPubMed
2.
3.
go back to reference Sahin AA, Ro JY, el-Naggar AK et al (1991) Tumor proliferative fraction in solid malignant neoplasms. A comparative study of Ki-67 immunostaining and flow cytometric determinations. Am J Clin Pathol 96(4):512–519CrossRefPubMed Sahin AA, Ro JY, el-Naggar AK et al (1991) Tumor proliferative fraction in solid malignant neoplasms. A comparative study of Ki-67 immunostaining and flow cytometric determinations. Am J Clin Pathol 96(4):512–519CrossRefPubMed
4.
go back to reference St Croix B, Flørenes VA, Rak JW et al (1996) Impact of the cyclin-dependent kinase inhibitor p27Kip1 on resistance of tumor cells to anticancer agents. Nat Med 2(11):1204–1210CrossRefPubMed St Croix B, Flørenes VA, Rak JW et al (1996) Impact of the cyclin-dependent kinase inhibitor p27Kip1 on resistance of tumor cells to anticancer agents. Nat Med 2(11):1204–1210CrossRefPubMed
5.
go back to reference Gardner LB, Li Q, Park MS, Flanagan WM, Semenza GL, Dang CV (2001) Hypoxia inhibits G1/S transition through regulation of p27 expression. J Biol Chem 276(11):7919–7926CrossRefPubMed Gardner LB, Li Q, Park MS, Flanagan WM, Semenza GL, Dang CV (2001) Hypoxia inhibits G1/S transition through regulation of p27 expression. J Biol Chem 276(11):7919–7926CrossRefPubMed
6.
7.
go back to reference Kim JJ, Tannock IF (2005) Repopulation of cancer cells during therapy: an important cause of treatment failure. Nat Rev Cancer 5(7):516–525CrossRefPubMed Kim JJ, Tannock IF (2005) Repopulation of cancer cells during therapy: an important cause of treatment failure. Nat Rev Cancer 5(7):516–525CrossRefPubMed
8.
go back to reference Brown JM (1999) The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture. Cancer Res 59(23):5863–5870PubMed Brown JM (1999) The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture. Cancer Res 59(23):5863–5870PubMed
9.
go back to reference Finch RA, Liu M, Grill SP et al (2000) Triapine (3-aminopyridine-2-carboxaldehyde- thiosemicarbazone): a potent inhibitor of ribonucleotide reductase activity with broad spectrum antitumor activity. Biochem Pharmacol 59(8):983–991CrossRefPubMed Finch RA, Liu M, Grill SP et al (2000) Triapine (3-aminopyridine-2-carboxaldehyde- thiosemicarbazone): a potent inhibitor of ribonucleotide reductase activity with broad spectrum antitumor activity. Biochem Pharmacol 59(8):983–991CrossRefPubMed
10.
go back to reference Richardson DR (2002) Iron chelators as therapeutic agents for the treatment of cancer. Crit Rev Oncol Hematol 42(3):267–281CrossRefPubMed Richardson DR (2002) Iron chelators as therapeutic agents for the treatment of cancer. Crit Rev Oncol Hematol 42(3):267–281CrossRefPubMed
11.
go back to reference Lovejoy DB, Jansson PJ, Brunk UT, Wong J, Ponka P, Richardson DR (2011) Antitumor activity of metal-chelating compound Dp44mT is mediated by formation of a redox-active copper complex that accumulates in lysosomes. Cancer Res 71(17):5871–5880CrossRefPubMed Lovejoy DB, Jansson PJ, Brunk UT, Wong J, Ponka P, Richardson DR (2011) Antitumor activity of metal-chelating compound Dp44mT is mediated by formation of a redox-active copper complex that accumulates in lysosomes. Cancer Res 71(17):5871–5880CrossRefPubMed
12.
go back to reference Shao J, Zhou B, Di Bilio AJ et al (2006) A ferrous-Triapine complex mediates formation of reactive oxygen species that inactivate human ribonucleotide reductase. Mol Cancer Ther 5(3):586–592CrossRefPubMed Shao J, Zhou B, Di Bilio AJ et al (2006) A ferrous-Triapine complex mediates formation of reactive oxygen species that inactivate human ribonucleotide reductase. Mol Cancer Ther 5(3):586–592CrossRefPubMed
13.
go back to reference Zhang X, Fryknäs M, Hernlund E et al (2014) Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments. Nat Commun 5:3295CrossRefPubMedPubMedCentral Zhang X, Fryknäs M, Hernlund E et al (2014) Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments. Nat Commun 5:3295CrossRefPubMedPubMedCentral
14.
go back to reference Eisenhauer EA, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45(2):228–247CrossRef Eisenhauer EA, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45(2):228–247CrossRef
15.
go back to reference Lawen A, Lane DJ (2013) Mammalian iron homeostasis in health and disease: uptake, storage, transport, and molecular mechanisms of action. Antioxid Redox Signal 18(18):2473–2507CrossRefPubMed Lawen A, Lane DJ (2013) Mammalian iron homeostasis in health and disease: uptake, storage, transport, and molecular mechanisms of action. Antioxid Redox Signal 18(18):2473–2507CrossRefPubMed
16.
go back to reference Merlot AM, Kalinowski DS, Richardson DR (2013) Novel chelators for cancer treatment: where are we now? Antioxid Redox Signal 18(8):973–1006CrossRefPubMed Merlot AM, Kalinowski DS, Richardson DR (2013) Novel chelators for cancer treatment: where are we now? Antioxid Redox Signal 18(8):973–1006CrossRefPubMed
17.
go back to reference Yu Y, Kovacevic Z, Richardson DR (2007) Tuning cell cycle regulation with an iron key. Cell Cycle 6(16):1982–1994CrossRefPubMed Yu Y, Kovacevic Z, Richardson DR (2007) Tuning cell cycle regulation with an iron key. Cell Cycle 6(16):1982–1994CrossRefPubMed
18.
go back to reference Le NT, Richardson DR (2002) The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochim Biophys Acta 1603(1):31–46PubMed Le NT, Richardson DR (2002) The role of iron in cell cycle progression and the proliferation of neoplastic cells. Biochim Biophys Acta 1603(1):31–46PubMed
19.
go back to reference Andrews NC (2000) Iron homeostasis: insights from genetics and animal models. Nat Rev Genet 1(3):208–217CrossRefPubMed Andrews NC (2000) Iron homeostasis: insights from genetics and animal models. Nat Rev Genet 1(3):208–217CrossRefPubMed
20.
go back to reference Yuan J, Lovejoy DB, Richardson DR (2004) Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood 104(5):1450–1458CrossRefPubMed Yuan J, Lovejoy DB, Richardson DR (2004) Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood 104(5):1450–1458CrossRefPubMed
21.
go back to reference Whitnall M, Howard J, Ponka P, Richardson DR (2006) A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc Natl Acad Sci U S A 103(40):14901–14906CrossRefPubMedPubMedCentral Whitnall M, Howard J, Ponka P, Richardson DR (2006) A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc Natl Acad Sci U S A 103(40):14901–14906CrossRefPubMedPubMedCentral
22.
go back to reference Defamie N, Chepied A, Mesnil M (2014) Connexins, gap junctions and tissue invasion. FEBS Lett 588(8):1331–1338CrossRefPubMed Defamie N, Chepied A, Mesnil M (2014) Connexins, gap junctions and tissue invasion. FEBS Lett 588(8):1331–1338CrossRefPubMed
23.
go back to reference Jansson PJ, Sharpe PC, Bernhardt PV, Richardson DR (2010) Novel thiosemicarbazones of the ApT and DpT series and their copper complexes: identification of pronounced redox activity and characterization of their antitumor activity. J Med Chem 53(15):5759–5769CrossRefPubMed Jansson PJ, Sharpe PC, Bernhardt PV, Richardson DR (2010) Novel thiosemicarbazones of the ApT and DpT series and their copper complexes: identification of pronounced redox activity and characterization of their antitumor activity. J Med Chem 53(15):5759–5769CrossRefPubMed
24.
go back to reference Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA (2007) Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada clinical trials group (NCIC IND.161). Investig New Drugs 25(5):471–477CrossRef Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA (2007) Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada clinical trials group (NCIC IND.161). Investig New Drugs 25(5):471–477CrossRef
25.
go back to reference Kunos CA, Winter K, Dicker AP et al (2013) Ribonucleotide reductase expression in cervical cancer: a radiation therapy oncology group translational science analysis. Int J Gynecol Cancer 23(4):615–621CrossRefPubMedPubMedCentral Kunos CA, Winter K, Dicker AP et al (2013) Ribonucleotide reductase expression in cervical cancer: a radiation therapy oncology group translational science analysis. Int J Gynecol Cancer 23(4):615–621CrossRefPubMedPubMedCentral
Metadata
Title
A phase I study of the safety and tolerability of VLX600, an Iron Chelator, in patients with refractory advanced solid tumors
Authors
Kabir Mody
Aaron S. Mansfield
Lalitha Vemireddy
Peter Nygren
Joachim Gulbo
Mitesh Borad
Publication date
01-08-2019
Publisher
Springer US
Published in
Investigational New Drugs / Issue 4/2019
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-018-0703-9

Other articles of this Issue 4/2019

Investigational New Drugs 4/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine