NAMI-A

Last updated
NAMI-A
NAMI-A.png
Clinical data
Routes of
administration
Intravenous
ATC code
  • None
Identifiers
  • Imidazolium-trans-tetrachloro(dimethylsulfoxide)imidazoleruthenium(III)
CAS Number
UNII
Chemical and physical data
Formula C8H15Cl4N4ORuS
Molar mass 458.17 g·mol−1

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. [1] [2] [3]

Contents

Reactions

NAMI-A is considered a pro-drug formulation that becomes active upon hydrolysis. At pH 7.4, a chloride is replaced by water giving a charge-neutral aquo complex. At lower pH, imidazole is cleaved and replaced with water. [4]

NAMI-A dissociates differently based on pH (Sava et al., 2001). NAMI-A pH.png
NAMI-A dissociates differently based on pH (Sava et al., 2001).

Unlike cisplatin, a platinum based drug, NAMI-A affects metastasis. [5] Whereas platinum compounds can be highly cytotoxic, NAMI-A is much less so. [6]

Preclinical trials

Several preclinical trials were conducted in various model systems.

In 2002, a study published by Vacca et al. showed that endothelial cells incubated with NAMI-A, were not able to proliferate but that NAMI-A did not kill the cells that were already grown. [7]

One trial was done to test the efficacy of NAMI-A in female [8] grafted with MCa mammary carcinoma cells. It was determined that, NAMI-A showed very little toxicity yet managed to decrease the rate of metastasis. Perhaps most impressively is that, though NAMI-A is very sensitive to the environment, it is able to be effective over a range of conditions. [4]

Clinical trials

Clinical trials were conducted at the Netherlands Cancer Institute (NKI).

Phase I – monotherapy

Phase I trials were conducted on NAMI-A in patients with varying solid tumors 3 hrs a day, 5 days a week, for 3 weeks at varying doses. Drug was given intravenously with and without a port-a-cath. Several side effects were observed including: [9]

Of the 24 patients in the study, 20 were evaluated for final results. At the end of the study, 19 of the 20 showed disease progression while 1 patient showed no progression. [9]

Due to these results, Phase II trials, using NAMI-A as a solo drug, were not pursued.[ citation needed ]

Phase I and II – combination therapy with gemcitabine

Due to negative results of the stand-alone Phase I trial, and knowledge that NAMI-A slows down progression of metastasis, and not growth of the initial tumor, Phase I & II trials were done using gemcitabine, a nucleoside analog that has shown to be successful in treating lung cancer.[ citation needed ]

Phase I trial

Phase I trials were done to determine the optimal and maximum tolerated dose (MTD)of NAMI-A as well as determining the pharmacokinetics of NAMI-A and gemcitabine administered jointly. This was accomplished by a dose escalation study. The minimal dosage was 300 mg/m2 on a 28-day schedule and the max dosage was 600 mg/m2 administered on a 21-day schedule. 32 patients were enrolled in the study all of which had a form of non-small cell lung cancer (NSCLC), a median age of 57, most diagnosed as level III & IV of disease progression. [9]

Phase II trial

Phase II trials were done to evaluate how NAMI-A in combination with gemcitabine impact cancer progression. 19 patients were added in addition to those from the phase I trial. Of the 27 patients evaluated for final results -15 showed anti-tumor activity, 10 showed stable disease progression for 6–10 weeks, and 1 patient exhibited a partial remission (PR) on the 300 mg/m2 for 21 days.[ citation needed ]

The patient who was observed to have a partial remission, occurred during the dose escalation phase I trial. In order to expand the trial, there needed to be at least one patient in the phase II trial that showed PR as the best response. Unfortunately, this did not occur. In addition, it was found that the results of NAMI-A in combination with gemcitabine, did not show improved results from studies done with gemcitabine alone. [9]

Due to these results, clinical trials were terminated.[ citation needed ]

Nomenclature

Contrary to what can be found in some papers, the nickname NAMI is not the acronym of “New Anticancer Metastasis Inhibitor”, but has a much more prosaic origin. It was created by a student as a short-form name of the chemical formula of the complex: “NA” comes from the symbol for sodium and “MI” from the word imidazole. The corresponding imidazolium salt was simply called NAMI-A to signify that it was an upgraded version of the prototype NAMI". [10]

Related Research Articles

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

Tirapazamine 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">Sunitinib</span> Cancer medication

Sunitinib, sold under the brand name Sutent, is an anti-cancer medication. It is a small-molecule, multi-targeted receptor tyrosine kinase (RTK) inhibitor that was approved by the FDA for the treatment of renal cell carcinoma (RCC) and imatinib-resistant gastrointestinal stromal tumor (GIST) in January 2006. Sunitinib was the first cancer drug simultaneously approved for two different indications.

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

Axitinib, sold under the brand name Inlyta, is a small molecule tyrosine kinase inhibitor developed by Pfizer. It has been shown to significantly inhibit growth of breast cancer in animal (xenograft) models and has shown partial responses in clinical trials with renal cell carcinoma (RCC) and several other tumour types.

Leronlimab is a humanized monoclonal antibody targeted against the CCR5 receptor found on T lymphocytes of the human immune system. It is being investigated as a potential therapy in the treatment of triple negative breast cancer and HIV infection.

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

Evofosfamide is a compound being evaluated in clinical trials for the treatment of multiple tumor types as a monotherapy and in combination with chemotherapeutic agents and other targeted cancer drugs.

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

Sapacitabine is a chemotherapeutic drug developed by US biotechnology firm Cyclacel currently undergoing clinical trials against leukemia.

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

Apatinib, also known as rivoceranib, is a tyrosine kinase inhibitor that selectively inhibits the vascular endothelial growth factor receptor-2. It is an orally bioavailable, small molecule agent which is thought to inhibit angiogenesis in cancer cells; specifically, apatinib inhibits VEGF-mediated endothelial cell migration and proliferation thus blocking new blood vessel formation in tumor tissue. This agent also mildly inhibits c-Kit and c-SRC tyrosine kinases.

Treatment of lung cancer refers to the use of medical therapies, such as surgery, radiation, chemotherapy, immunotherapy, percutaneous ablation, and palliative care, alone or in combination, in an attempt to cure or lessen the adverse impact of malignant neoplasms originating in lung tissue.

Lipoplatin is a nanoparticle of 110 nm average diameter composed of lipids and cisplatin. This new drug has successfully finished Phase I, Phase II, and Phase III human clinical trials. It has shown superiority to cisplatin in combination with paclitaxel as a chemotherapy regimen in non-small cell lung cancer (NSCLC) adenocarcinomas.

<span class="mw-page-title-main">Temozolomide</span> Cancer medication

Temozolomide, sold under the brand name Temodar among others, is an anticancer medication used to treat brain tumors such as glioblastoma and anaplastic astrocytoma. It is taken by mouth or via intravenous infusion.

Angiokinase inhibitors are a new therapeutic target for the management of cancer. They inhibit tumour angiogenesis, one of the key processes leading to invasion and metastasis of solid tumours, by targeting receptor tyrosine kinases. Examples include nintedanib, afatinib and motesanib.

Demcizumab is a humanized monoclonal antibody which is used to treat patients with pancreatic cancer or non-small cell lung cancer. Demcizumab has completed phase 1 trials and is currently undergoing phase 2 trials. Demcizumab was developed by OncoMed Pharmaceuticals in collaboration with Celgene.

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.

Arabinopyranosyl-<i>N</i>-methyl-<i>N</i>-nitrosourea Chemical compound

Arabinopyranosyl-N-methyl-N-nitrosourea, also known as Aranose (Араноза) is a cytostatic anticancer chemotherapeutic drug of an alkylating type. Chemically it is a nitrosourea derivative. It was developed in the Soviet Union in the 1970s. It was claimed by its developers that its advantages over other nitrosoureas are a relatively low hematological toxicity and a wider therapeutic index, which allows for its outpatient administration.

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

Crisnatol (BW-A770U) is an experimental anticancer agent known for its potential in inhibiting the growth of various solid tumors. Research has indicated that crisnatol acts as a DNA-intercalating agent, thereby disrupting the replication process in cancer cells. A Phase I clinical trial was conducted to assess its safety profile, pharmacokinetics, and potential efficacy in patients with solid malignancies. This study highlighted the drug’s ability to inhibit tumor growth, although associated toxicities were observed, necessitating further research to optimize its therapeutic window.

<span class="mw-page-title-main">KP1019</span> Chemical compound

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.

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

Capmatinib, sold under the brand name Tabrecta, is an anticancer medication used for the treatment of metastatic non-small cell lung cancer whose tumors have a mutation that leads to the exon 14 skipping of the MET gene, which codes for the membrane receptor HGFR.

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 February 2024, BOLD-100 was being tested in a Phase 1b/2a clinical trial in 117 patients with advanced gastrointestinal cancers in combination with the chemotherapy regimen FOLFOX. BOLD-100 is being developed by Bold Therapeutics Inc.

<span class="mw-page-title-main">Rebimastat</span> Investigational antineoplastic drug

Rebimastat is an abandoned investigational antineoplastic drug developed as a broad-spectrum matrix metalloproteinase inhibitor (MMPI). It was designed to target enzymes implicated in cancer progression, aiming to reduce tumor growth and metastasis. Although promising in preclinical studies, clinical development was halted due to adverse effects.

References

  1. Kostova I (2006). "Ruthenium complexes as anticancer agents". Current Medicinal Chemistry. 13 (9): 1085–1107. doi:10.2174/092986706776360941. PMID   16611086.
  2. Lentz F, Drescher A, Lindauer A, Henke M, Hilger RA, Hartinger CG, et al. (February 2009). "Pharmacokinetics of a novel anticancer ruthenium complex (KP1019, FFC14A) in a phase I dose-escalation study". Anti-Cancer Drugs. 20 (2): 97–103. doi:10.1097/CAD.0b013e328322fbc5. PMID   19209025. S2CID   38457574.
  3. Alessio E, Messori L (2018). "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?". In Sigel A, Sigel H, Freisinger E, Sigel RK (eds.). Metallo-Drugs:Development and Action of Anticancer Agents. Metal Ions in Life Sciences. Vol. 18. Berlin: de Gruyter GmbH. pp. 141–170. doi:10.1515/9783110470734-011. ISBN   9783110470734. PMID   29394024.
  4. 1 2 Sava G, Bergamo A, Zorzet S, Gava B, Casarsa C, Cocchietto M, et al. (February 2002). "Influence of chemical stability on the activity of the antimetastasis ruthenium compound NAMI-A". European Journal of Cancer. 38 (3): 427–435. doi:10.1016/S0959-8049(01)00389-6. PMID   11818210.
  5. Sava G, Zorzet S, Turrin C, Vita F, Soranzo M, Zabucchi G, et al. (May 2003). "Dual Action of NAMI-A in inhibition of solid tumor metastasis: selective targeting of metastatic cells and binding to collagen" (PDF). Clinical Cancer Research. 9 (5): 1898–1905. PMID   12738748.
  6. Bergamo A, Gagliardi R, Scarcia V, Furlani A, Alessio E, Mestroni G, Sava G (April 1999). "In vitro cell cycle arrest, in vivo action on solid metastasizing tumors, and host toxicity of the antimetastatic drug NAMI-A and cisplatin" (PDF). The Journal of Pharmacology and Experimental Therapeutics. 289 (1): 559–564. PMID   10087050.
  7. Vacca A, Bruno M, Boccarelli A, Coluccia M, Ribatti D, Bergamo A, et al. (March 2002). "Inhibition of endothelial cell functions and of angiogenesis by the metastasis inhibitor NAMI-A". British Journal of Cancer. 86 (6): 993–998. doi:10.1038/sj.bjc.6600176. PMC   2364145 . PMID   11953835.
  8. Lukashevich IS, Malkovský M, Schorlemmer HU, Bobé P, Artym J. "Mice, Inbred CBA". WikiGenes.
  9. 1 2 3 4 Leijen S, Burgers SA, Baas P, Pluim D, Tibben M, van Werkhoven E, et al. (February 2015). "Phase I/II study with ruthenium compound NAMI-A and gemcitabine in patients with non-small cell lung cancer after first line therapy". Investigational New Drugs. 33 (1): 201–214. doi:10.1007/s10637-014-0179-1. hdl: 1874/314465 . PMID   25344453. S2CID   27817700.
  10. Alessio E, Guo Z (September 28, 2016). "Thirty Years of the Drug Candidate NAMI-A and the Myths in the Field of Ruthenium Anticancer Compounds: A Personal Perspective". European Journal of Inorganic Chemistry. 2017 (12): 1549–1560. doi:10.1002/ejic.201600986. hdl: 11368/2898222 .