Cisplatin

Last updated

Cisplatin
Cisplatin-stereo.svg
Cisplatin-3D-balls.png Cisplatin-3D-vdW.png
Clinical data
Trade names Platinol, others
Other namesCisplatinum, platamin, neoplatin, cismaplat, cis-diamminedichloroplatinum(II) (CDDP)
AHFS/Drugs.com Monograph
MedlinePlus a684036
License data
Pregnancy
category
Routes of
administration
Intravenous
ATC code
Legal status
Legal status
Pharmacokinetic data
Bioavailability 100% (IV)
Protein binding > 95%
Elimination half-life 30–100 hours
Excretion Renal
Identifiers
  • (SP-4-2)-diamminedichloridoplatinum(II)
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
PDB ligand
CompTox Dashboard (EPA)
ECHA InfoCard 100.036.106 OOjs UI icon edit-ltr-progressive.svg
Chemical and physical data
Formula [Pt(NH3)2Cl2]
Molar mass 300.05 g·mol−1
3D model (JSmol)
  • [NH3+]-[Pt-2](Cl)(Cl)[NH3+]
  • InChI=1S/2ClH.2H3N.Pt/h2*1H;2*1H3;/q;;;;+2/p-2 Yes check.svgY
  • Key:LXZZYRPGZAFOLE-UHFFFAOYSA-L Yes check.svgY
 X mark.svgNYes check.svgY  (what is this?)    (verify)

Cisplatin is a chemical compound with formula cis-[Pt(NH3)2Cl2]. It is a coordination complex of platinum that is used as a chemotherapy medication used to treat a number of cancers. [3] These include testicular cancer, ovarian cancer, cervical cancer, bladder cancer, head and neck cancer, esophageal cancer, lung cancer, mesothelioma, brain tumors and neuroblastoma. [3] It is given by injection into a vein. [3]

Contents

Common side effects include bone marrow suppression, hearing problems including severe hearing loss, kidney damage, and vomiting. [3] [4] [5] Other serious side effects include numbness, trouble walking, allergic reactions, electrolyte problems, and heart disease. [3] Use during pregnancy can cause harm to the developing fetus. [1] [3] Cisplatin is in the platinum-based antineoplastic family of medications. [3] It works in part by binding to DNA and inhibiting its replication. [3]

Cisplatin was discovered in 1845 and licensed for medical use in 1978 and 1979. [6] [3] It is on the World Health Organization's List of Essential Medicines. [7] [8]

Medical use

Cisplatin is administered intravenously as short-term infusion in normal saline for treatment of solid and haematological malignancies. It is used to treat various types of cancers, including sarcomas, some carcinomas (e.g., small cell lung cancer, squamous cell carcinoma of the head and neck and ovarian cancer), lymphomas, bladder cancer, cervical cancer, [9] and germ cell tumors.

The introduction of cisplatin as a standard treatment for testicular cancer improved remission rates from 5-10% before 1974 to 75-85% by 1984. [10]

Side effects

Cisplatin has a number of side effects that can limit its use:

Pharmacology

Cisplatin interferes with DNA replication, which kills the fastest proliferating cells, which in theory are cancerous. Following administration, one chloride ion is slowly displaced by water to give the aquo complex cis-[PtCl(NH3)2(H2O)]+, in a process termed aquation. Dissociation of the chloride is favored inside the cell because the intracellular chloride concentration is only 3–20% of the approximately 100 mM chloride concentration in the extracellular fluid. [21] [22]

The water molecule in cis-[PtCl(NH3)2(H2O)]+ is itself easily displaced by the N-heterocyclic bases on DNA. Guanine preferentially binds. A model compound has been prepared and crystals were examined by X-ray crystallography [23]

Subsequent to formation of [PtCl(guanine-DNA)(NH3)2]+, crosslinking can occur via displacement of the other chloride, typically by another guanine. [24] Cisplatin crosslinks DNA in several different ways, interfering with cell division by mitosis. The damaged DNA elicits DNA repair mechanisms, which in turn activate apoptosis when repair proves impossible. In 2008, researchers were able to show that the apoptosis induced by cisplatin on human colon cancer cells depends on the mitochondrial serine-protease Omi/Htra2. [25] Since this was only demonstrated for colon carcinoma cells, it remains an open question whether the Omi/Htra2 protein participates in the cisplatin-induced apoptosis in carcinomas from other tissues. [25]

Most notable among the changes in DNA are the 1,2-intrastrand cross-links with purine bases. These include 1,2-intrastrand d(GpG) adducts, which form nearly 90% of the adducts, and the less common 1,2-intrastrand d(ApG) adducts. Coordination chemists have obtained crystals of the products of reacting cisplain with small models of DNA. Here is a POVray plot of the platinum binding to a small model of DNA. [26]

A POVray plot of the atomic coordinates for the cis Pt(NH3)2 and short fragment of DNA which was reported by Stephen J. Lippard in Science 1985 Cisplain adduct with two Gs bonded to ribose and linked by phosphate.png
A POVray plot of the atomic coordinates for the cis Pt(NH3)2 and short fragment of DNA which was reported by Stephen J. Lippard in Science 1985

1,3-intrastrand d(GpXpG) adducts occur but are readily excised by the nucleotide excision repair (NER). Other adducts include inter-strand crosslinks and nonfunctional adducts that have been postulated to contribute to cisplatin's activity. Interaction with cellular proteins, particularly HMG domain proteins, has also been advanced as a mechanism of interfering with mitosis, although this is probably not its primary method of action. [27]

Cisplatin resistance

Cisplatin combination chemotherapy is the cornerstone of treatment of many cancers. Initial platinum responsiveness is high, but the majority of cancer patients will eventually relapse with cisplatin-resistant disease. Many mechanisms of cisplatin resistance have been proposed, including changes in cellular uptake and efflux of the drug, increased detoxification of the drug, inhibition of apoptosis , increased DNA repair or changes in metabolism. [28] [29] Oxaliplatin is active in highly cisplatin-resistant cancer cells in the laboratory; however, there is little evidence for its activity in the clinical treatment of patients with cisplatin-resistant cancer. [29] The drug paclitaxel may be useful in the treatment of cisplatin-resistant cancer; the mechanism for this activity is as yet unknown. [30]

Transplatin

Transplatin, the trans-stereoisomer of cisplatin, has formula trans-[PtCl2(NH3)2] and does not exhibit a comparably useful pharmacological effect. Two mechanisms have been suggested to explain the reduced anticancer effect of transplatin. Firstly, the trans arrangement of the chloro ligands is thought to confer transplatin with greater chemical reactivity, causing transplatin to become deactivated before it reaches the DNA, where cisplatin exerts its pharmacological action. Secondly, the stereo-conformation of transplatin is such that it is unable to form the characteristic 1,2-intrastrand d(GpG) adducts formed by cisplatin in abundance. [31]

Molecular structure

Cisplatin is the square planar coordination complex cis-[Pt(NH3)2Cl2]. [32] :286–8 [33] :689 The prefix cis indicates the cis isomer in which two similar ligands are in adjacent positions. [32] [33] :550 The systematic chemical name of this molecule is cis–diamminedichloroplatinum, [32] :286 where ammine with two m's indicates an ammonia (NH3) ligand, as opposed to an organic amine with one m. [32] :284

History

The compound cis-[Pt(NH3)2Cl2] was first described by Italian chemist Michele Peyrone in 1845, and known for a long time as Peyrone's salt. [34] [35] The structure was deduced by Alfred Werner in 1893. [24] In 1965, Barnett Rosenberg, Van Camp et al. of Michigan State University discovered that electrolysis of platinum electrodes generated a soluble platinum complex which inhibited binary fission in Escherichia coli (E. coli) bacteria. Although bacterial cell growth continued, cell division was arrested, the bacteria growing as filaments up to 300 times their normal length. [36] The octahedral Pt(IV) complex cis-[PtCl4(NH3)2], but not the trans isomer, was found to be effective at forcing filamentous growth of E. coli cells. The square planar Pt(II) complex, cis-[PtCl2(NH3)2] turned out to be even more effective at forcing filamentous growth. [37] [38] This finding led to the observation that cis-[PtCl2(NH3)2] was indeed highly effective at regressing the mass of sarcomas in rats. [39] Confirmation of this discovery, and extension of testing to other tumour cell lines launched the medicinal applications of cisplatin. Cisplatin was approved for use in testicular and ovarian cancers by the U.S. Food and Drug Administration on 19 December 1978. [24] [40] [41] and in the UK (and in several other European countries) in 1979. [42] Cisplatin was the first to be developed. [43] In 1983 pediatric oncologist Roger Packer began incorporating cisplatin into adjuvant chemotherapy for the treatment of childhood medulloblastoma. [44] The new protocol that he developed led to a marked increase in disease-free survival rates for patients with medulloblastoma, up to around 85%. [45] The Packer Protocol has since become a standard treatment for medulloblastoma. Likewise, cisplatin has been found to be particularly effective against testicular cancer, where its use improved the cure rate from 10% to 85%. [10]

Recently, some researchers have investigated at the preclinical level new forms of cisplatin prodrugs in combination with nanomaterials in order to localize the release of the drug in the target. [46] [47]

Synthesis

Syntheses of cisplatin start from potassium tetrachloroplatinate. Several procedures are available. One obstacle is the facile formation of Magnus's green salt (MGS), which has the same empirical formula as cisplatin. The traditional way to avoid MGS involves the conversion of K2PtCl4 to K2PtI4, as originally described by Dhara. [48] [49] Reaction with ammonia forms PtI2(NH3)2 which is isolated as a yellow compound. When silver nitrate in water is added insoluble silver iodide precipitates and [Pt(OH2)2(NH3)2](NO3)2 remains in solution. Addition of potassium chloride will form the final product which precipitates [49] In the triiodo intermediate the addition of the second ammonia ligand is governed by the trans effect. [49]

Cisplatin synthesis.svg

A one-pot synthesis of cisplatin from K2PtCl4 has been developed. It relies on the slow release of ammonia from ammonium acetate. [50]

Research

Cisplatin has been studied with Auger therapy to increase the therapeutic effects of cisplatin, without increasing normal tissue toxicities. [51] However, due to significant side effects, the search for structurally novel Pt(II) and Pd(II) compounds exhibiting antineoplastic activity is extremely important and aims to develop more effective and less toxic drugs. [52] Metal complexes comprising cisplatin-like molecules ([PtCl(NH3)2] or [Pt(NH3)Cl2]) linked by variable length alkandiamine chains have attracted great interest in the last few years as next-generation alternatives drugs in cancer chemotherapy. [53] [54] [55]

Related Research Articles

<span class="mw-page-title-main">Carboplatin</span> Medication used to treat cancer

Carboplatin, sold under the brand name Paraplatin among others, is a chemotherapy medication used to treat a number of forms of cancer. This includes ovarian cancer, lung cancer, head and neck cancer, brain cancer, and neuroblastoma. It is used by injection into a vein.

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

Oxaliplatin, sold under the brand name Eloxatin among others, is a cancer medication used to treat colorectal cancer. It is given by injection into a vein.

<span class="mw-page-title-main">Metal ammine complex</span>

In coordination chemistry, metal ammine complexes are metal complexes containing at least one ammonia ligand. "Ammine" is spelled this way for historical reasons; in contrast, alkyl or aryl bearing ligands are spelt with a single "m". Almost all metal ions bind ammonia as a ligand, but the most prevalent examples of ammine complexes are for Cr(III), Co(III), Ni(II), Cu(II) as well as several platinum group metals.

<span class="mw-page-title-main">Non-small-cell lung cancer</span> Any type of epithelial lung cancer other than small-cell lung carcinoma

Non-small-cell lung cancer (NSCLC), or non-small-cell lung carcinoma, is any type of epithelial lung cancer other than small-cell lung cancer (SCLC). NSCLC accounts for about 85% of all lung cancers. As a class, NSCLCs are relatively insensitive to chemotherapy, compared to small-cell carcinoma. When possible, they are primarily treated by surgical resection with curative intent, although chemotherapy has been used increasingly both preoperatively and postoperatively.

Triplatin tetranitrate is a platinum-based cytotoxic drug that underwent clinical trials for the treatment of human cancer. The drug acts by forming adducts with cellular DNA, preventing DNA transcription and replication, thereby inducing apoptosis. Other platinum-containing anticancer drugs include cisplatin, carboplatin, and oxaliplatin.

<span class="mw-page-title-main">Crosslinking of DNA</span> Phenomenon in genetics

In genetics, crosslinking of DNA occurs when various exogenous or endogenous agents react with two nucleotides of DNA, forming a covalent linkage between them. This crosslink can occur within the same strand (intrastrand) or between opposite strands of double-stranded DNA (interstrand). These adducts interfere with cellular metabolism, such as DNA replication and transcription, triggering cell death. These crosslinks can, however, be repaired through excision or recombination pathways.

<span class="mw-page-title-main">Alkylating antineoplastic agent</span> Pharmaceutical drugs

An alkylating antineoplastic agent is an alkylating agent used in cancer treatment that attaches an alkyl group (CnH2n+1) to DNA.

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

Potassium tetrachloroplatinate(II) is the chemical compound with the formula K2PtCl4. This reddish orange salt is an important reagent for the preparation of other coordination complexes of platinum. It consists of potassium cations and the square planar dianion PtCl42−. Related salts are also known including Na2PtCl4, which is brown-colored and soluble in alcohols, and quaternary ammonium salts, which are soluble in a broader range of organic solvents.

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

Satraplatin is a platinum-based antineoplastic agent that was under investigation as a treatment of patients with advanced prostate cancer who have failed previous chemotherapy. It has not yet received approval from the U.S. Food and Drug Administration. First mentioned in the medical literature in 1993, satraplatin is the first orally active platinum-based chemotherapeutic drug; other available platinum analogues—cisplatin, carboplatin, and oxaliplatin—must be given intravenously.

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

Azasetron is an antiemetic which acts as a 5-HT3 receptor antagonist, pKi = 9.27 It is used in the management of nausea and vomiting induced by cancer chemotherapy (such as cisplatin chemotherapy). Azasetron hydrochloride is given in a usual dose of 10 mg once daily by mouth or intravenously. It is approved for marketing in Japan, and marketed exclusively by Torii Pharmaceutical Co., Ltd. under the trade names "Serotone I.V. Injection 10 mg" and "Serotone Tablets 10 mg". Pharmacokinetics data from S. Tsukagoshi.

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.

<span class="mw-page-title-main">Stephen J. Lippard</span> American chemist

Stephen James Lippard is the Arthur Amos Noyes Emeritus Professor of Chemistry at the Massachusetts Institute of Technology. He is considered one of the founders of bioinorganic chemistry, studying the interactions of nonliving substances such as metals with biological systems. He is also considered a founder of metalloneurochemistry, the study of metal ions and their effects in the brain and nervous system. He has done pioneering work in understanding protein structure and synthesis, the enzymatic functions of methane monooxygenase (MMO), and the mechanisms of cisplatin anticancer drugs. His work has applications for the treatment of cancer, for bioremediation of the environment, and for the development of synthetic methanol-based fuels.

Platinum-based antineoplastic drugs are chemotherapeutic agents used to treat cancer. Their active moieties are coordination complexes of platinum. These drugs are used to treat almost half of people receiving chemotherapy for cancer. In this form of chemotherapy, commonly used drugs include cisplatin, oxaliplatin, and carboplatin, but several have been proposed or are under development. Addition of platinum-based chemotherapy drugs to chemoradiation in women with early cervical cancer seems to improve survival and reduce risk of recurrence.

Auger therapy is a form of radiation therapy for the treatment of cancer which relies on low-energy electrons to damage cancer cells, rather than the high-energy radiation used in traditional radiation therapy. Similar to other forms of radiation therapy, Auger therapy relies on radiation-induced damage to cancer cells to arrest cell division, stop tumor growth and metastasis and kill cancerous cells. It differs from other types of radiation therapy in that electrons emitted via the Auger effect are released with low kinetic energy. In contrast to traditional α- and β-particle emitters, Auger electron emitters exhibit low cellular toxicity during transit in blood or bone marrow.

Antineoplastic resistance, often used interchangeably with chemotherapy resistance, is the resistance of neoplastic (cancerous) cells, or the ability of cancer cells to survive and grow despite anti-cancer therapies. In some cases, cancers can evolve resistance to multiple drugs, called multiple drug resistance.

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

Phenanthriplatin or cis-[Pt(NH3)2-(phenanthridine)Cl]NO3 is a new drug candidate. It belongs to a family of platinum(II)-based agents which includes cisplatin, oxaliplatin and carboplatin. Phenanthriplatin was discovered by Professor Stephen J. Lippard at Massachusetts Institute of Technology and is currently being developed by Blend Therapeutics for its potential use in human cancer therapy.

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

Dicycloplatin is a chemotherapy medication used to treat a number of cancers which includes the non-small-cell lung carcinoma and prostate cancer.

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

trans-Dichlorodiammineplatinum(II) is the trans isomer of the coordination complex with the formula trans-PtCl2(NH3)2, sometimes called transplatin. It is a yellow solid with low solubility in water but good solubility in DMF. The existence of two isomers of PtCl2(NH3)2 led Alfred Werner to propose square planar molecular geometry. It belongs to the molecular symmetry point group D2h.

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">Ototoxic medication</span>

Ototoxicity is defined as the toxic effect on the functioning of the inner ear, which may lead to temporary or permanent hearing loss (cochleotoxic) and balancing problems (vestibulotoxic). Drugs or pharmaceutical agents inducing ototoxicity are regarded as ototoxic medications.

References

  1. 1 2 3 "Cisplatin Use During Pregnancy". Drugs.com. 12 September 2019. Retrieved 25 February 2020.
  2. "FDA-sourced list of all drugs with black box warnings (Use Download Full Results and View Query links.)". nctr-crs.fda.gov. FDA . Retrieved 22 October 2023.
  3. 1 2 3 4 5 6 7 8 9 "Cisplatin". The American Society of Health-System Pharmacists. Archived from the original on 21 December 2016. Retrieved 8 December 2016.
  4. Oun R, Moussa YE, Wheate NJ (May 2018). "The side effects of platinum-based chemotherapy drugs: a review for chemists". Dalton Transactions. 47 (19): 6645–6653. doi:10.1039/c8dt00838h. PMID   29632935.
  5. 1 2 3 Callejo A, Sedó-Cabezón L, Juan ID, Llorens J (July 2015). "Cisplatin-Induced Ototoxicity: Effects, Mechanisms and Protection Strategies". Toxics. 3 (3): 268–293. doi: 10.3390/toxics3030268 . PMC   5606684 . PMID   29051464.
  6. Fischer J, Ganellin CR (2006). Analogue-based Drug Discovery. John Wiley & Sons. p. 513. ISBN   9783527607495. Archived from the original on 20 December 2016.
  7. World Health Organization (2019). World Health Organization model list of essential medicines: 21st list 2019. Geneva: World Health Organization. hdl: 10665/325771 . WHO/MVP/EMP/IAU/2019.06. License: CC BY-NC-SA 3.0 IGO.
  8. World Health Organization (2021). World Health Organization model list of essential medicines: 22nd list (2021). Geneva: World Health Organization. hdl: 10665/345533 . WHO/MHP/HPS/EML/2021.02.
  9. "Cisplatin". National Cancer Institute. 2 March 2007. Archived from the original on 8 October 2014. Retrieved 13 November 2014.
  10. 1 2 Einhorn LH (November 1990). "Treatment of testicular cancer: a new and improved model". Journal of Clinical Oncology. 8 (11): 1777–81. doi:10.1200/JCO.1990.8.11.1777. PMID   1700077.
  11. 1 2 Miller RP, Tadagavadi RK, Ramesh G, Reeves WB (October 2010). "Mechanisms of Cisplatin Nephrotoxicity". Toxins. 2 (11): 2490–2518. doi: 10.3390/toxins2112490 . PMC   3153174 . PMID   22069563.
  12. Singh N, Vik A, Lybrand DB, Morisseau C, Hammock BD (November 2021). "New Alkoxy- Analogues of Epoxyeicosatrienoic Acids Attenuate Cisplatin Nephrotoxicity In Vitro via Reduction of Mitochondrial Dysfunction, Oxidative Stress, Mitogen-Activated Protein Kinase Signaling, and Caspase Activation". Chemical Research in Toxicology. 34 (12): 2579–2591. doi:10.1021/acs.chemrestox.1c00347. PMC   8853703 . PMID   34817988.
  13. 1 2 3 Milosavljevic N, Duranton C, Djerbi N, Puech PH, Gounon P, Lagadic-Gossmann D, et al. (October 2010). "Nongenomic effects of cisplatin: acute inhibition of mechanosensitive transporters and channels without actin remodeling". Cancer Research. 70 (19): 7514–22. doi: 10.1158/0008-5472.CAN-10-1253 . PMID   20841472.
  14. Squillace S, Niehoff ML, Doyle TM, Green M, Esposito E, Cuzzocrea S, Arnatt CK, Spiegel S, Farr SA, Salvemini D (September 2022). "Sphingosine-1-phosphate receptor 1 activation in the central nervous system drives cisplatin-induced cognitive impairment". The Journal of Clinical Investigation. 132 (17). doi:10.1172/JCI157738. PMC   9433103 . PMID   36047496.
  15. "Unlocking the Mystery of "Chemo Brain"". Neuroscience News. 2 September 2022.
  16. Orgel E, Villaluna D, Krailo MD, Esbenshade A, Sung L, Freyer DR (May 2022). "Sodium thiosulfate for prevention of cisplatin-induced hearing loss: updated survival from ACCL0431". The Lancet. Oncology. 23 (5): 570–572. doi:10.1016/S1470-2045(22)00155-3. PMC   9635495 . PMID   35489339.
  17. Winstead E (6 October 2022). "Sodium Thiosulfate Reduces Hearing Loss in Kids with Cancer". National Cancer Institute. Retrieved 9 March 2023.
  18. "FDA approves sodium thiosulfate to reduce the risk of ototoxicity associated with cisplatin in pediatric patients with localized, non-metastatic solid tumors". Food and Drug Administration. 20 September 2022. Retrieved 9 March 2023.
  19. Sarafraz Z, Ahmadi A, Daneshi A (June 2018). "Transtympanic Injections of N-acetylcysteine and Dexamethasone for Prevention of Cisplatin-Induced Ototoxicity: Double Blind Randomized Clinical Trial". The International Tinnitus Journal. 22 (1): 40–45. doi:10.5935/0946-5448.20180007. PMID   29993216.
  20. Levi JA, Aroney RS, Dalley DN (June 1981). "Haemolytic anaemia after cisplatin treatment". British Medical Journal. 282 (6281): 2003–4. doi:10.1136/bmj.282.6281.2003. PMC   1505958 . PMID   6788166.
  21. Wang D, Lippard SJ (April 2005). "Cellular processing of platinum anticancer drugs". Nature Reviews. Drug Discovery. 4 (4): 307–320. doi:10.1038/nrd1691. PMID   15789122. S2CID   31357727.
  22. Johnstone TC, Suntharalingam K, Lippard SJ (March 2016). "The Next Generation of Platinum Drugs: Targeted Pt(II) Agents, Nanoparticle Delivery, and Pt(IV) Prodrugs". Chemical Reviews. 116 (5): 3436–3486. doi:10.1021/acs.chemrev.5b00597. PMC   4792284 . PMID   26865551.
  23. Orbell JD, Solorzano C, Marzilli LG, Kistenmacher TJ (October 1982). "Preparation and structure of cis-chlorodiammine (N2, N2-dimethyl-9-methylguanine) platinum (II) hexafluorophosphate. A model for the intermediate in the proposed crosslinking mode of action of platinum (II) antitumor agents". Inorganic Chemistry. 21 (10): 3806–3810. doi:10.1021/ic00140a041.
  24. 1 2 3 Trzaska S (20 June 2005). "Cisplatin". Chemical & Engineering News . 83 (25): 52. doi:10.1021/cen-v083n025.p052.
  25. 1 2 Pruefer FG, Lizarraga F, Maldonado V, Melendez-Zajgla J (June 2008). "Participation of Omi Htra2 serine-protease activity in the apoptosis induced by cisplatin on SW480 colon cancer cells". Journal of Chemotherapy. 20 (3): 348–354. doi:10.1179/joc.2008.20.3.348. PMID   18606591. S2CID   11052459.
  26. Sherman SE, Gibson D, Wang AH, Lippard SJ (October 1985). "X-ray structure of the major adduct of the anticancer drug cisplatin with DNA: cis-[Pt(NH3)2(d(pGpG))]". Science. 230 (4724): 412–7. Bibcode:1985Sci...230..412S. doi:10.1126/science.4048939. PMID   4048939.
  27. Hu J, Lieb JD, Sancar A, Adar S (October 2016). "Cisplatin DNA damage and repair maps of the human genome at single-nucleotide resolution". PNAS. 113 (41): 11507–11512. Bibcode:2016PNAS..11311507H. doi: 10.1073/pnas.1614430113 . PMC   5068337 . PMID   27688757. S2CID   11052459.
  28. Cruz-Bermúdez A, Laza-Briviesca R, Vicente-Blanco RJ, García-Grande A, Coronado MJ, Laine-Menéndez S, et al. (May 2019). "Cisplatin resistance involves a metabolic reprogramming through ROS and PGC-1α in NSCLC which can be overcome by OXPHOS inhibition". Free Radical Biology & Medicine. 135: 167–181. doi:10.1016/j.freeradbiomed.2019.03.009. hdl: 10486/688357 . PMID   30880247.
  29. 1 2 Stordal B, Davey M (November 2007). "Understanding cisplatin resistance using cellular models" (PDF). IUBMB Life. 59 (11): 696–699. doi: 10.1080/15216540701636287 . PMID   17885832. S2CID   30879019.
  30. Stordal B, Pavlakis N, Davey R (December 2007). "A systematic review of platinum and taxane resistance from bench to clinic: an inverse relationship" (PDF). Cancer Treatment Reviews. 33 (8): 688–703. doi:10.1016/j.ctrv.2007.07.013. hdl:2123/4068. PMID   17881133.
  31. Coluccia M, Natile G (January 2007). "Trans-platinum complexes in cancer therapy". Anti-Cancer Agents in Medicinal Chemistry. 7 (1): 111–123. doi:10.2174/187152007779314080. PMID   17266508.
  32. 1 2 3 4 Miessler GL, Tarr DA (1999). Inorganic Chemistry (2nd ed.). Prentice Hall. ISBN   978-0-13-841891-5.
  33. 1 2 Housecroft CE, Sharpe AG (2005). Inorganic Chemistry (2nd ed.). Pearson Prentice Hall. ISBN   978-0-130-39913-7.
  34. Kauffman GB, Pentimalli R, Hall MD (2010). "Michele Peyrone (1813–1883), Discoverer of Cisplatin". Platinum Metals Review. 54 (4): 250–256. doi: 10.1595/147106710X534326 . Retrieved 3 October 2022. This biographical article aims to present, for the first time in the English language, a summary of his life and the achievements that he made during his scientific career.
  35. Peyrone M (1844). "Ueber die Einwirkung des Ammoniaks auf Platinchlorür" [On the action of ammonia on platinum chloride]. Ann. Chem. Pharm. 51 (1): 1–29. doi:10.1002/jlac.18440510102.
  36. Rosenberg B, Vancamp L, Krigas T (February 1965). "Inhibition of cell division in Escherichia coli by electrolysis products from a platinum electrode". Nature. 205 (4972): 698–9. Bibcode:1965Natur.205..698R. doi:10.1038/205698a0. PMID   14287410. S2CID   9543916.
  37. Rosenberg B, Van Camp L, Grimley EB, Thomson AJ (March 1967). "The inhibition of growth or cell division in Escherichia coli by different ionic species of platinum(IV) complexes". The Journal of Biological Chemistry. 242 (6): 1347–52. doi: 10.1016/S0021-9258(18)96186-7 . PMID   5337590.
  38. Christie DA, Tansey EM (2007). Christie DA, Tansey EM, Thomson AJ (eds.). The Discovery, Use and Impact of Platinum Salts as Chemotherapy Agent for Cancer. Wellcome Trust Witnesses to Twentieth Century Medicine. Vol. 30. pp. 6–15. ISBN   978-0-85484-112-7.
  39. Rosenberg B, VanCamp L, Trosko JE, Mansour VH (April 1969). "Platinum compounds: a new class of potent antitumour agents". Nature. 222 (5191): 385–6. Bibcode:1969Natur.222..385R. doi:10.1038/222385a0. PMID   5782119. S2CID   32398470.
  40. Carpenter DP (2010). Reputation and power: organizational image and pharmaceutical regulation at the FDA. Princeton, NJ: Princeton University Press. ISBN   978-0-691-14180-0.
  41. "Approval Summary for cisplatin for Metastatic ovarian tumors". FDA Oncology Tools. Food and Drug Administration, Center for Drug Evaluation and Research. 19 December 1978. Archived from the original on 8 February 2008. Retrieved 15 July 2009.
  42. Wiltshaw E (1979). "Cisplatin in the treatment of cancer". Platinum Metals Review. 23 (3): 90–8. doi:10.1595/003214079X2339098. S2CID   267470502.
  43. Kelland L (2007). "The resurgence of platinum-based cancer chemotherapy". Nature Reviews Cancer. 7 (8): 573–584. doi:10.1038/nrc2167. PMID   17625587. S2CID   205468214.
  44. Packer RJ, Sutton LN, Elterman R, Lange B, Goldwein J, Nicholson HS, et al. (November 1994). "Outcome for children with medulloblastoma treated with radiation and cisplatin, CCNU, and vincristine chemotherapy". Journal of Neurosurgery. 81 (5): 690–8. doi:10.3171/jns.1994.81.5.0690. PMID   7931615.
  45. Packer RJ, Sutton LN, Goldwein JW, Perilongo G, Bunin G, Ryan J, et al. (March 1991). "Improved survival with the use of adjuvant chemotherapy in the treatment of medulloblastoma". Journal of Neurosurgery. 74 (3): 433–40. doi:10.3171/jns.1991.74.3.0433. PMID   1847194.
  46. Dhar S, Daniel WL, Giljohann DA, Mirkin CA, Lippard SJ (October 2009). "Polyvalent oligonucleotide gold nanoparticle conjugates as delivery vehicles for platinum(IV) warheads". Journal of the American Chemical Society. 131 (41): 14652–3. doi:10.1021/ja9071282. PMC   2761975 . PMID   19778015.
  47. Santi M, Mapanao AK, Cassano D, Vlamidis Y, Cappello V, Voliani V (April 2020). "Endogenously-Activated Ultrasmall-in-Nano Therapeutics: Assessment on 3D Head and Neck Squamous Cell Carcinomas". Cancers. 12 (5): 1063. doi: 10.3390/cancers12051063 . PMC   7281743 . PMID   32344838.
  48. Dhara SC (1970). "Cisplatin". Indian J. Chem. 8: 123–134.
  49. 1 2 3 Alderden RA, Hall MD, Hambley TW (2006). "The Discovery and Development of Cisplatin". J. Chem. Educ. 83 (5): 728. Bibcode:2006JChEd..83..728A. doi:10.1021/ed083p728. S2CID   29546931.
  50. Kukushikin VY, Oskarsson Å, Elding LI, Farrell N (2007). "Facile Synthesis of Isomerically Pure cis -Dichlorodiammineplatinum(II), Cisplatin". Facile Synthesis of Isomerically Pure cis -Dichlorodiammineplatinum(II), Cisplatin. Inorganic Syntheses. Vol. 32. pp. 141–144. doi:10.1002/9780470132630.ch23. ISBN   9780470132630.
  51. Ku A, Facca VJ, Cai Z, Reilly RM (October 2019). "Auger electrons for cancer therapy - a review". EJNMMI Radiopharmacy and Chemistry. 4 (1): 27. doi: 10.1186/s41181-019-0075-2 . PMC   6800417 . PMID   31659527.
  52. Fiuza SM, Amado AM, Oliveira PJ, Sardão VA, De Carvalho LB, Marques MP (2006). "Pt(II) vs Pd(II) Polyamine Complexes as New Anticancer Drugs: A Structure- Activity Study". Letters in Drug Design & Discovery. 3 (3): 149–151. doi:10.2174/157018006776286989. hdl: 10316/45139 .
  53. Teixeira LJ, Seabra M, Reis E, da Cruz MT, de Lima MC, Pereira E, et al. (May 2004). "Cytotoxic activity of metal complexes of biogenic polyamines: polynuclear platinum(II) chelates". Journal of Medicinal Chemistry. 47 (11): 2917–2925. doi:10.1021/jm0311238. hdl: 10316/10605 . PMID   15139770.
  54. Vinci D, Chateigner D (1 December 2022). "Synthesis and structural characterization of a new dinuclear platinum(III) complex, [Pt 2 Cl 4 (NH 3 ) 2 {μ-HN=C(O)Bu t } 2 ]". Acta Crystallographica Section B Structural Science, Crystal Engineering and Materials. 78 (6): 835–841. doi:10.1107/S2052520622009660. ISSN   2052-5206. PMC   9728019 .
  55. Omondi RO, Ojwach SO, Jaganyi D (November 2020). "Review of comparative studies of cytotoxic activities of Pt(II), Pd(II), Ru(II)/(III) and Au(III) complexes, their kinetics of ligand substitution reactions and DNA/BSA interactions". Inorganica Chimica Acta. 512: 119883. doi:10.1016/j.ica.2020.119883. S2CID   225575546.

Further reading