KP1019

Last updated
KP1019
KP1019.tif
KP1019 Chemical Structure
Clinical data
Routes of
administration
Intravenous (as a sodium salt, KP1339)
Identifiers
  • trans-[Tetrachlorobis(1H-indazole)ruthenate(III)]
UNII
ChEBI
Chemical and physical data
Formula C21H19Cl4N6Ru
Molar mass 598.29 g·mol−1

KP1019, or indazole trans-[tetrachlorobis(1H-indazole)ruthenate(III)], is one of four ruthenium anti-cancer drugs to enter into phase I clinical trials, the others being BOLD-100, NAMI-A and TLD-1433. Research into ruthenium-based drugs has provided novel alternatives for platinum-based chemotherapeutics such as Cisplatin and its derivatives.  KP1019 is useful for metastatic tumors and cis-platin resistant tumors.  It exhibits potent cytotoxicity against primary tumors, particularly in colorectal cancer. [1]

Contents

Structure and properties

KP1019 has an octahedral structure with two trans N-donor indazole and four chloride ligands in the equatorial plane.  It has a low solubility in water, which makes it difficult to transport in the bloodstream. The Ru-Cl bonds are labile and KP1019 readily exchanges its chloro ligands in the presence of water. [2]

KP1019 derivatives

Due to its low solubility in water, KP1019 is often prepared as its sodium salt, the basis for KP-1339 and BOLD-100.  By replacing the indazole rings with imidazole rings the derivative KP418 is formed.  KP418 also exhibits anti-cancer activity, however it has not completed Phase I clinical testing.  KP418 exhibits slower cellular uptake and slower protein binding.  Similar to KP418, replacement of one of the imidazole ligands with DMSO yields NAMI-A.  NAMI-A is considered to be one of two leading ruthenium-based anti-cancer drugs, along with BOLD-100.  Both have entered clinical trials. [1]

Synthesis

KP1019 is synthesized by refluxing RuCl3-3H2O with HCl and ethanol.  The ethanol is removed and indazole is allowed to react with the solution at 70 °C.  The resulting solid is collected by filtration and its purity is evaluate by UV-visible spectroscopy, elemental analysis, and determination of reduction potential. [3]

Mechanism of action

Human Transferrin (Tf) PDB: 2HAU Transferrin.2HAU.jpg
Human Transferrin (Tf) PDB: 2HAU

Activation by reduction

The active form of KP1019 has Ru in its 2+ state. The hypoxic environment of cancer cell tissue facilitates this reduction and the specific action on cancer cells over healthy cells. The intracellular reducing agent is unknown, but glutathione is a good candidate as it reduces promiscuously and has a reduction potential on par with the transition of Ru from 3+ to 2+. This mechanism of action is favorable in terms of efficacy as well. An increase in Ru (III) reduction potential positively correlates with the complex's antiproliferative activity. [1]

Human Serum Albumin (HSA) PDB: 1AO6 HSA.1AO6.jpg
Human Serum Albumin (HSA) PDB: 1AO6

Reactivity with serum proteins

KP1019 binds transferrin (Tf), a 700 amino acid glycoprotein, in the pocket usually bound to 2 atoms of Fe3+. The transferrin protein binds to the transferrin receptor and is taken into the cell by endocytosis.  This protein and its receptors are overexpressed in cancer cells owing to their increased demand for iron, and it is believed that Tf transport is the reason ruthenium compounds accumulate in tumors. CD and ESI-MS studies have shown that one molecule of Tf binds two ruthenium complexes via. The intracellular release requires a significant increase in pH due to the high binding affinity. Citric acid or adenosine 5’-triphosphate, which are both present in vivo, are capable of liberating KP1019. [4]

Human serum albumin, the most present protein in blood plasma, binds to KP1019 in a 1:4 protein:drug ratio. In plasma, it is almost exclusively bound to protein, up to 90%. It is possible that albumin serves to bind available Ru drugs until they are transported into the cell by Tf. [5]

Interaction with DNA

Most metal-based anti-tumor compounds interact strongly with DNA.  Binding assays of KP1019 with the four common nucleotide bases reveal a preference for guanosine 5’-monophosphate and adenosine 5’-monophosphate.  KP1019 is able to untwist and bend DNA weakly.  While Pt-based compounds also target purines, KP1019's DNA lesions may differ in quantity and strength.  In tumor cells, the drug induces 15-fold lower interstrand DNA cross-linking efficiency than cisplatin.  The interaction of KP1019 and its imidazole-containing analogue KP418 with DNA increases in the hypoxic environment that tumor cells are subject to.  This correspondingly increased cytotoxicity as well. [1]

Preclinical cancer efficacy

KP1019 and KP1339 both induce apoptosis in colorectal tumor cell lines SW480 and HT29.  This is induced predominantly by a loss of mitochondrial membrane potential in a high percentage of cells. DNA strand breaks are not believed to be the cause of the major damage.  This is due to KP1019-induced death being independent of the cell's p53 status.  Studies have shown oxidative stress to contribute towards KP1019-induced apoptosis.  Furthermore, KP1019 has been shown to counteract tumor resistance to other metal-based drugs. KP1019 does not appear to be susceptible to the same cancer cell mechanism of acquiring drug resistance as other metal-based drugs, and remains potent in cell-lines known for their drug resistance. [6] KP1019 has significant antineoplastic activity in chemically induced colorectal carcinoma in rats surpassing its in-vitro activity. The rat model of colorectal carcinoma is highly analogous to human colorectal carcinoma and presents a promising target for the drug. [7] [8] The yeast strain Saccharomyces cerevisiae serves as a cellular model for KP1019 and manifests induced DNA damage, cell cycle delay, and cell death. [3]

Phase I clinical trials

KP1019 was administered to patients twice weekly in doses ranging from 25 mg to 600 mg for three weeks. Patients had advanced solid tumors. Five out of six patients saw disease stabilization for up to 10 weeks. No serious side effects of KP1019 were reported. [1]

Related Research Articles

<span class="mw-page-title-main">Transferrin</span> Mammalian protein found in Homo sapiens

Transferrins are glycoproteins found in vertebrates which bind and consequently mediate the transport of iron (Fe) through blood plasma. They are produced in the liver and contain binding sites for two Fe3+ ions. Human transferrin is encoded by the TF gene and produced as a 76 kDa glycoprotein.

<span class="mw-page-title-main">Cisplatin</span> Pharmaceutical drug

Cisplatin is a chemotherapy medication used to treat a number of cancers. These include testicular cancer, ovarian cancer, cervical cancer, bladder cancer, head and neck cancer, esophageal cancer, lung cancer, mesothelioma, brain tumors and neuroblastoma. It is given by injection into a vein.

<span class="mw-page-title-main">Cancer immunotherapy</span> Artificial stimulation of the immune system to treat cancer

Cancer immunotherapy is the stimulation of the immune system to treat cancer, improving on the immune system's natural ability to fight the disease. It is an application of the fundamental research of cancer immunology and a growing subspecialty of oncology.

<span class="mw-page-title-main">Targeted therapy</span> Type of therapy

Targeted therapy or molecularly targeted therapy is one of the major modalities of medical treatment (pharmacotherapy) for cancer, others being hormonal therapy and cytotoxic chemotherapy. As a form of molecular medicine, targeted therapy blocks the growth of cancer cells by interfering with specific targeted molecules needed for carcinogenesis and tumor growth, rather than by simply interfering with all rapidly dividing cells. Because most agents for targeted therapy are biopharmaceuticals, the term biologic therapy is sometimes synonymous with targeted therapy when used in the context of cancer therapy. However, the modalities can be combined; antibody-drug conjugates combine biologic and cytotoxic mechanisms into one targeted therapy.

<span class="mw-page-title-main">Tirapazamine</span> Chemical compound

Tirapazamine (SR-[[4233]]) is an experimental anticancer drug that is activated to a toxic radical only at very low levels of oxygen (hypoxia). Such levels are common in human solid tumors, a phenomenon known as tumor hypoxia. Thus, tirapazamine is activated to its toxic form preferentially in the hypoxic areas of solid tumors. Cells in these regions are resistant to killing by radiotherapy and most anticancer drugs. Thus the combination of tirapazamine with conventional anticancer treatments is particularly effective. As of 2006, tirapazamine is undergoing phase III testing in patients with head and neck cancer and gynecological cancer, and similar trials are being undertaken for other solid tumor types.

<span class="mw-page-title-main">Mechanism of action</span> Biochemical interaction through which a drug produces its pharmacological effect

In pharmacology, the term mechanism of action (MOA) refers to the specific biochemical interaction through which a drug substance produces its pharmacological effect. A mechanism of action usually includes mention of the specific molecular targets to which the drug binds, such as an enzyme or receptor. Receptor sites have specific affinities for drugs based on the chemical structure of the drug, as well as the specific action that occurs there.

<span class="mw-page-title-main">Betulinic acid</span> Chemical compound

Betulinic acid is a naturally occurring pentacyclic triterpenoid which has antiretroviral, antimalarial, and anti-inflammatory properties, as well as a more recently discovered potential as an anticancer agent, by inhibition of topoisomerase. It is found in the bark of several species of plants, principally the white birch from which it gets its name, but also the ber tree, selfheal, the tropical carnivorous plants Triphyophyllum peltatum and Ancistrocladus heyneanus, Diospyros leucomelas, a member of the persimmon family, Tetracera boiviniana, the jambul, flowering quince, rosemary, and Pulsatilla chinensis.

A metastasis suppressor is a protein that acts to slow or prevent metastases from spreading in the body of an organism with cancer. Metastasis is one of the most lethal cancer processes. This process is responsible for about ninety percent of human cancer deaths. Proteins that act to slow or prevent metastases are different from those that act to suppress tumor growth. Genes for about a dozen such proteins are known in humans and other animals.

<span class="mw-page-title-main">PINX1</span> Protein-coding gene in the species Homo sapiens

PIN2/TERF1-interacting telomerase inhibitor 1, also known as PINX1, is a human gene. PINX1 is also known as PIN2 interacting protein 1. PINX1 is a telomerase inhibitor and a possible tumor suppressor.

<span class="mw-page-title-main">Carmofur</span> Chemical compound

Carmofur (INN) or HCFU (1-hexylcarbamoyl-5-fluorouracil) is a pyrimidine analogue used as an antineoplastic agent. It is a derivative of fluorouracil, being a lipophilic-masked analog of 5-FU that can be administered orally.

<span class="mw-page-title-main">Cancer cell</span> Tumor cell

Cancer cells are cells that divide continually, forming solid tumors or flooding the blood or lymph with abnormal cells. Cell division is a normal process used by the body for growth and repair. A parent cell divides to form two daughter cells, and these daughter cells are used to build new tissue or to replace cells that have died because of aging or damage. Healthy cells stop dividing when there is no longer a need for more daughter cells, but cancer cells continue to produce copies. They are also able to spread from one part of the body to another in a process known as metastasis.

<span class="mw-page-title-main">NAMI-A</span> Chemical compound

NAMI-A is the imidazolium]] salt of the coordination complex [RuCl4(dmso)(C3N2H4)] where dmso is dimethylsulfoxide and C3N2H4 is imidazole Together with KP1019 and BOLD-100, NAMI-A has been investigated as an anticancer agent.

When overexpressed ectopically, anticancer genes preferentially kill cancer cells while sparing the normal healthy cells. Apoptosis, apoptosis following a mitotic catastrophe, necrosis, and autophagy are only a few of the processes that can lead to cell death. In the late 1990s, research on cancer cells led to the identification of anticancer genes. Currently, '291 The human genome' contains anti-cancer genes. Base substitutions that lead to insertions, deletions, or alterations in missense amino acids cause frameshifts altering the protein. The gene codes for copy number variations or gene rearrangements lead to their deregulation are necessary for a gene change in copy number or gene rearrangements. (1)

<span class="mw-page-title-main">Cancer epigenetics</span> Field of study in cancer research

Cancer epigenetics is the study of epigenetic modifications to the DNA of cancer cells that do not involve a change in the nucleotide sequence, but instead involve a change in the way the genetic code is expressed. Epigenetic mechanisms are necessary to maintain normal sequences of tissue specific gene expression and are crucial for normal development. They may be just as important, if not even more important, than genetic mutations in a cell's transformation to cancer. The disturbance of epigenetic processes in cancers, can lead to a loss of expression of genes that occurs about 10 times more frequently by transcription silencing than by mutations. As Vogelstein et al. points out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations. However, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 heavily methylated CpG islands in the promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa. Manipulation of epigenetic alterations holds great promise for cancer prevention, detection, and therapy. In different types of cancer, a variety of epigenetic mechanisms can be perturbed, such as the silencing of tumor suppressor genes and activation of oncogenes by altered CpG island methylation patterns, histone modifications, and dysregulation of DNA binding proteins. There are several medications which have epigenetic impact, that are now used in a number of these diseases.

Ruthenium anti-cancer drugs are coordination complexes of ruthenium complexes that have anticancer properties. They promise to provide alternatives to platinum-based drugs for anticancer therapy. No ruthenium anti-cancer drug has been commercialized.

<span class="mw-page-title-main">Titanocene Y</span>

Titanocene Y also known as bis[(p-methoxybenzyl)cyclopentadienyl]titanium(IV) dichloride or dichloridobis(η5-(p-methoxybenzyl)cyclopentadienyl)titanium is an organotitanium compound that has been investigated for use as an anticancer drug.

<span class="mw-page-title-main">Christian Hartinger</span> New Zealand bioinorganic chemist known for his work in metal-based anticancer drugs

Christian G. Hartinger is an Austrian-born New Zealand bioinorganic chemist known for his work in metal-based anticancer drugs. In 2022 he was elected a Fellow of the Royal Society Te Apārangi.

VEGFR-2 inhibitor, also known as kinase insert domain receptor(KDR) inhibitor, are tyrosine kinase receptor inhibitors that reduce angiogenesis or lymphangiogenesis, leading to anticancer activity. Generally they are small, synthesised molecules that bind competitively to the ATP-site of the tyrosine kinase domain. VEGFR-2 selective inhibitor can interrupt multiple signaling pathways involved in tumor, including proliferation, metastasis and angiogenesis.

RAPTA is a class of experimental cancer drugs. They consist of a central ruthenium(II) atom complexed to an arene group, chlorides, and 1,3,5-triaza-7-phosphaadamantane (PTA) forming an organoruthenium half-sandwich compound. Other related ruthenium anti-cancer drugs include NAMI-A, KP1019 and BOLD-100.

<span class="mw-page-title-main">BOLD-100</span> Experimental cancer drug

BOLD-100, or sodium trans-[tetrachlorobis (1H-indazole)ruthenate(III)], is a ruthenium-based anti-cancer therapeutic in clinical development. As of November 2021, BOLD-100 was being tested in a Phase 1b clinical trial in patients with advanced gastrointestinal cancers in combination with the chemotherapy regimen FOLFOX. BOLD-100 is being developed by Bold Therapeutics Inc.

References

  1. 1 2 3 4 5 Hartinger CG, Jakupec MA, Zorbas-Seifried S, Groessl M, Egger A, Berger W, Zorbas H, Dyson PJ, Keppler BK (October 2008). "KP1019, a new redox-active anticancer agent--preclinical development and results of a clinical phase I study in tumor patients". Chemistry & Biodiversity. 5 (10): 2140–55. doi:10.1002/cbdv.200890195. PMID   18972504. S2CID   205544866.
  2. Farrell NP, Gorle AK, Peterson EJ, Berners-Price SJ (February 2018). Sigel A, Sigel H, Freisinger E, Sigel RK (eds.). "Chapter 5. The Deceptively Similar Ruthenium(III) Drug Candidates KP1019 and NAMI-A have Different Actions. What Did We Learn in the Past 30 Years?". Metal Ions in Life Sciences. de Gruyter GmbH. 18: 141–170. doi:10.1515/9783110470734-010. PMID   29394023.
  3. 1 2 Stevens SK, Strehle AP, Miller RL, Gammons SH, Hoffman KJ, McCarty JT, Miller ME, Stultz LK, Hanson PK (January 2013). "The anticancer ruthenium complex KP1019 induces DNA damage, leading to cell cycle delay and cell death in Saccharomyces cerevisiae". Molecular Pharmacology. 83 (1): 225–34. doi:10.1124/mol.112.079657. PMID   23090979. S2CID   33746600.
  4. Pongratz M, Schluga P, Jakupec MA, Arion VB, Hartinger CG, Allmaier G, Keppler BK (2004). "Transferrin binding and transferrin-mediated cellular uptake of the ruthenium coordination compound KP1019, studied by means of AAS, ESI-MS and CD spectroscopy". Journal of Analytical Atomic Spectrometry. 19 (1): 46–51. doi:10.1039/B309160K.
  5. Trynda-Lemiesz L, Karaczyn A, Keppler BK, Kozłowski H (March 2000). "Studies on the interactions between human serum albumin and trans-indazolium (bisindazole) tetrachlororuthenate(III)". Journal of Inorganic Biochemistry. 78 (4): 341–6. doi:10.1016/s0162-0134(00)00062-3. PMID   10857915.
  6. Heffeter P, Pongratz M, Steiner E, Chiba P, Jakupec MA, Elbling L, Marian B, Körner W, Sevelda F, Micksche M, Keppler BK, Berger W (January 2005). "Intrinsic and acquired forms of resistance against the anticancer ruthenium compound KP1019 [indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (FFC14A)". The Journal of Pharmacology and Experimental Therapeutics. 312 (1): 281–9. doi:10.1124/jpet.104.073395. PMID   15331656. S2CID   14212220.
  7. Seelig MH, Berger MR, Keppler BK (1992). "Antineoplastic activity of three ruthenium derivatives against chemically induced colorectal carcinoma in rats". Journal of Cancer Research and Clinical Oncology. 118 (3): 195–200. doi:10.1007/bf01410134. PMID   1548284. S2CID   25316271.
  8. Berger MR, Garzon FT, Keppler BK, Schmähl D (1989). "Efficacy of new ruthenium complexes against chemically induced autochthonous colorectal carcinoma in rats". Anticancer Research. 9 (3): 761–5. PMID   2764521.