Review article

NOWOTWORY Journal of Oncology

2022, volume 72, number 2, 96–105

DOI: 10.5603/NJO.a2022.0011

© Polskie Towarzystwo Onkologiczne

ISSN 0029–540X, e-ISSN: 2300-2115

www.nowotwory.edu.pl

Selected platinum complexes in standard and modern anti-cancer therapies

Anna Kopacz-BednarskaTeodora Król
Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Kielce, Poland
The constantly observed increase in the number of cancer cases inspires research aimed at searching for new compounds with anti-cancer potential. In recent years, much research has focused on platinum complexes, especially their anti-cancer properties. Platinum derivatives are characterized by high cytotoxic activity against many types of cancer cells. However, among the numerous developed complexes, only cisplatin, carboplatin and oxaliplatin have found wide application in chemotherapeutic treatment. Nedaplatin, lobaplatin and heptaplatin have also gained recognition, and have been implemented in oncological therapy in Japan, China and Korea. Some of the platinum complexes are still at the stage of pre-clinical and clinical trials. The aim of the research conducted today is to search for platinum compounds that show high pharmacological effectiveness, with clearly limited side effects. In future therapeutic strategies, the possibility of using platinum complexes in conjunction with other chemotherapeutic compounds is being considered, which may contribute to increasing the efficacy of anti-cancer therapy.
Key words: cisplatin, platinum complexes, anticancer drugs, chemotherapy

How to cite:

Kopacz-Bednarska A, Król T. Selected platinum complexes in standard and modern anti-cancer therapies. NOWOTWORY J Oncol 2022; 72: 96–105.

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially.

Introduction

According to the National Cancer Registry, cancer diseases, along with cardiovascular diseases, are the most common cause of death in Poland [1, 2]. Literature data show that the number of patients with neoplastic diseases may systematically increase, and cancer may become the main cause of premature deaths, for both women and men [3]. The most frequent cases of cancer are lung, ovarian, cervical, prostate, testicular, stomach and colon cancers. In addition to many currently used methods of treating oncological diseases, it is important to implement appropriate preventive measures in everyday life, which would significantly slow down the processes of carcinogenesis.

The etiology of neoplastic diseases is complex and multifaceted, conditioned by both external (environmental) and internal factors [4]. It has been shown that some behavioral and psychosocial factors (including stress and depression) as well as genetic predispositions may contribute to the development and progression of neoplastic diseases [5–7]. An improper diet, low physical activity and chronic stress are more and more often mentioned as some of the basic indicators influencing the development of the carcinogenesis process [5–7, 8, 9].

Despite the wide range of preventive tests implemented, the development of diagnostic techniques and the constantly growing public awareness, it has still not been possible to find appropriate therapeutic methods that would effectively combat all types of cancer. In recent years, special attention has been paid to the side effects of treatment, resulting from the high toxicity of the cytostatics used [10, 11]. It was also noticed that the problem of cancer cells resistance to the drugs used is becoming more and more frequent [12, 13]. Numerous metal compounds, including platinum and its derivatives, play an important role in the treatment of neoplastic diseases. Currently, some platinum complexes are used effectively in the chemotherapy of malignant neoplasms. However, there are still some platinum derivatives whose anti-tumor activity is not yet sufficiently known and described.

The aim of this study is to explain the most important mechanisms of action of the selected platinum compounds, their potential therapeutic properties, and to determine the role of new platinum complexes that would be characterized by low toxicity over a broad spectrum of anti-tumor activity.

Platinum complexes in anti-cancer therapy

Due to the multidirectional scope of action of platinum, platinum drugs are now quite widely used in the treatment of cancer. One of them is the platinum compound (II) called cisplatin. It was introduced to clinical practice at the earliest, in the literature it is described as a first-generation platinum drug. The confirmation of the anti-cancer effect of cisplatin has resulted in the search for other platinum complexes, effective in anti-cancer therapy, but with limited side effects [14]. In recent years, a number of in vitro and in vivo studies have been conducted to determine the potential anti-tumor properties of cisplatin analogues. Of these, only carboplatin and oxaliplatin are used in oncological therapy, and a number of others (picoplatin, phenanthriplatin, satraplatin, adamplatin, oxoplatin, ethacraplatin, lipoplatin, BBR3464) are still at the experimental or clinical trial stage (tab. I). The mechanisms underlying the anti-tumor activity of the new platinum (II) and (IV) complexes are still insufficiently elucidated. It is known, however, that platinum compounds are characterized by quite diverse therapeutic effects, which may result from a different chemical structure, geometric isomerism and the degree of oxidation of platinum [15].

Cisplatin – a first-generation platinum drug

Cisplatin was first synthesized by Alfred Werner in 1845, and its chemical structure was described in 1893. In 1965, Barnett Rosenberg showed that platinum complexes generated during electrolysis significantly weaken the multiplication of Escherichia coli bacteria [14, 16]. This discovery became the basis for further research aimed at determining the inhibitory effect of cisplatin on the proliferation of cancer cells. It was then revealed that the compound can effectively inhibit cell division of murine sarcoma and L1210 leukemia [16]. In clinical practice, cisplatin was first used in 1971, while 7 years later this compound was approved by the Food and Drug Administration (FDA) and became an available drug with an anti-cancer effect [17–19]. Currently, cisplatin is used with great effectiveness in the treatment of breast, ovarian, cervical, prostate, testicular, esophagus, stomach, head and neck cancer, multiple myeloma, melanoma, non-Hodgkin’s lymphoma and cell lung cancer [17, 18, 20].

The drug can be used both as monotherapy and in combination therapy with radiotherapy, taxoids (paclitaxel and docetaxel), doxorubicin, 5-fluorouracil, leucovorin and gemcitabine. The combined effects of cisplatin and other compounds in the treatment of various types of cancer are still undergoing numerous experimental and clinical evaluations [21–23]. Cisplatin has been shown to be highly effective in the treatment of neoplastic diseases, but at the same time it has been found to be highly toxic to normal cells. Side effects are multi-organ, include cardiotoxicity, ototoxicity, myelosuppressive and immunosuppressive activity [17, 24–26]. Moreover, cisplatin is a highly nephrotoxic drug leading to the development of acute renal failure, which may significantly impede dosing of the drug and limit its use [17, 27, 28].

Cisplatin is a cytostatic, belonging to the group of drugs with an alkylating effect [13]. It has pro-apoptotic [20, 29–31] and antiproliferative [20, 32, 33] properties, which allows it to be used in the treatment of many types of malignant neoplasms. However, it is important to remember about the factors influencing the effectiveness of cisplatin treatment, such as: the diverse biological response of cancer cells, various sensitivity and resistance to the drug. Neoplastic cell resistance to cisplatin may lead to disease recurrences, sometimes shortly after chemotherapy has been completed.

The mechanisms underlying platinum resistance are complex and are currently not fully understood. This process is multifactorial in nature. In general, several signals are activated simultaneously, which weaken the effectiveness of the therapy [33]. This is a key problem in overcoming the resistance of cancer cells to cisplatin. Therefore, it is extremely important to conduct research that will allow an explanation of the interaction between the factors, responsible for both the sensitivity and resistance, of cancer cells to the action of platinum complexes.

Cisplatin analogues currently used in cancer chemotherapy

The available literature shows that cisplatin is a long-used anticancer drug showing high toxicity and numerous dose-dependent side effects [17, 32–36]. In addition, acquired resistance to this drug has been found [33–35]. These data inspired the search for new compounds with cisplatin-like properties but with high therapeutic efficacy and limited adverse effects on normal cells. The platinum derivatives, carboplatin and oxaliplatin, turned out to be drugs with a broad spectrum of antitumor activity, with low toxicity and reduced acquired resistance. In addition to carboplatin and oxaliplatin, which have been approved and introduced in medicine, the platinum complexes such as nedaplatin, lobaplatin and heptaplatin are also gaining recognition. To date, only a few Asian countries have obtained consent to use these compounds in oncological therapy (tab. I).

Table I. Selected platinum complexes in contemporary and future therapeutic strategies [18, 36, 37, 71, 77, 96]. Current status as of July 2021

Platinum complex

Molecular formuła

Structure

Clinical or experimental status

Platinum complex

Molecular formuła

Structure

Clinical or experimental status

PLATINUM COMPLEXES (II)

cisplatin

platinol

cis-dichlorodiammine platinum

Pt(NH3)2Cl2

The drug was approved by the FDA. It has been used in medicine since 1978.

carboplatin paraplatin

cis-diammine(1.1-cyclobutanedicarboxylato) platinum

[C4H6(CO2)2]Pt(NH3)2

The drug was approved by the FDA. It has been used in medicine since 1989.

oxaliplatin

eloxatin

(trans-R,R-cyclohexane-1.2-diammine)oxalatoplatinum

[SP-4-2-(1R-trans)]-(1.2-cyclohexanediamine-N,N')[ethanedioata(2--)-O,O’]platinum
(DACH)PtCl2

C8H14N2O4Pt

The drug was approved by the FDA. It has been used in Europe since 1999 and in the USA since 2002.

nedaplatin

aqupla

cis-diammine(glycolato)platinum

C2H8N2O3Pt

The drug has been used in Japan since 1995. It is still the subject of numerous clinical trials.

lobaplatin

D-19466

1,2-diammino-1-methyl-cyclobutane-platinum-lactate

C9H18N2O3Pt

The drug has been used in China since 2004. It is still the subject of numerous clinical trials..

heptaplatin

SKI-2053R

sunpla

cis-malonato[(4R,5R)-4.5-bis(aminomethyl)-2-isopropyl-1.3-dioxolane]platinum

C11H22N2O6Pt

The drug has been used in Korea since 2005..

picoplatin

AMD473

JM473

ZD0473

cis-diammine-dichloro
(2-methylpyridine)platinum

C6H10Cl2N2Pt

The compound is in clinical trials.

phenanthriplatin

(SP-4-3)- diamminechlorido(phenanthridine)platinum nitrate,

cis-Pt(NH3)2(phenanthridine)Cl]NO3

cis-[Pt(NH3)2Cl(phenanthridine)]+

C13H15ClN4O3Pt

The compound is at the stage of experimental research..

PLATINUM
COMPLEXES (IV)

satraplatin

JM216

bis-acetato-amminedichloro

(cyclohexylamine) platinum

C10H22Cl2N2O4Pt

The compound is in clinical trials.

adamplatin

LA-12

trans-[PtCl2(CH3COO)2(NH3)
(1-adamantylamine)]

C14H26Cl2N2O4Pt

The compound is at the stage of experimental research.

oxoplatin

cis-diammine-dichlorido-trans-dihydroxy-platinum
Cl2H8N2O2Pt

Pt(NH3)2Cl2(OH)2

The compound is at the stage of experimental research.

ethacraplatin

cis,cis,trans-diamminodichloridobis(ethacrynato)platinum

The compound is at the stage of experimental research.

PLATINUM COMPLEX PACKED IN LIPOSOMES

lipoplatin

liposomal cisplatin

encapsulated

cis-diammine-dichloroplatinum(II)

The compound is in clinical trials.

TRI-NUCLEAR PLATINUM COMPLEX

BBR3464

triplatin tetranitrate

((trans-PtCl(NH3)2)2-(trans-Pt(NH3)2 (NH2(CH2)6NH2)2)NO3)4

C12H50Cl2N14O12Pt3

The compound is in clinical trials.

Carboplatin

Carboplatin is a second generation platinum compound with a structure similar to cisplatin. In the structure of carboplatin, instead of chlorine atoms, there is a 1.1-cyclobutyl dicarboxylic anion [36–38]. After penetrating the cell membrane, this compound is hydrolyzed to active forms, this process is much slower than the hydrolysis of cisplatin, therefore carboplatin may be better tolerated by patients. Through covalent bonds with N7 purine atoms, it forms adducts with DNA analogous to cisplatin, so that the mechanism of its biological activity is similar to that of cisplatin [25, 39]. The binding of carboplatin to the DNA of neoplastic cells leads to changes in the nucleic acid structure and inhibition of the replication process. This results in the induction of apoptosis and determines the cytotoxic properties of the compound [40]. The kinetics of carboplatin binding to DNA, however, is much slower than that of cisplatin. The slow hydrolysis of carboplatin shows, depending on the type of cancer, reduced effectiveness of the therapeutic effect compared to the effect of cisplatin [18, 41]. Literature data show that in order to obtain a cytotoxic effect similar to cisplatin, it is necessary to use up to 10 times higher doses of carboplatin [42]. Currently, carboplatin is used in the treatment of ovarian cancer of epithelial origin and non-small-cell lung cancer, and it is also administered in multi-drug therapy in the case of insufficient tolerance of the organism to cisplatin treatment [43]. The use of carboplatin in combination with taxoids and vincristine has also been suggested in the treatment of malignant tumors of the testicle, head and neck, cervical and breast cancer, and malignant glioma [39, 44–46]. Despite the lower pharmacological efficacy of carboplatin, a positive aspect of its use is its reduced systemic toxicity, especially its nephrotoxicity. On the other hand, an adverse effect demonstrated with carboplatin therapy is bone marrow dysfunction [47].

Oxaliplatin

Oxaliplatin is a platinum drug of the 3rd generation, the structure of which incorporates, in place of the amine ligands, a 1.2-diaminocyclohexane (DACH) group, which determines the cytotoxic activity of this complex [48]. This drug inhibits DNA repair processes, leading to inhibition of the cell cycle and increasing the sensitivity of cancer cells to signals from the apoptotic pathway [49]. Oxaliplatin exhibits a biological effect similar to first and second generation platinum drugs, consisting in the formation of specific adducts with DNA, interfering with replication and transcription of the deoxyribonucleic acid double helix [50]. As a result of oxaliplatin biotransformation, a secondary metabolite other than for cisplatin and carboplatin (trans-diaminacyclohexane-dihydroxy-platinum [II]) is formed, which may result in a different pharmacokinetic profile of the drug and various toxic effects [43, 48]. Oxaliplatin has been shown to have a relatively low activity in monotherapy, so it is most often administered in combination with other chemotherapeutic compounds (e.g., fluorouracil, irinotecan, leucovorin, ifosfamide, etoposide and gemcitabine) [48, 51]. Currently, this drug is used mainly in the multi-drug therapy of testicular, stomach and pancreatic cancer, breast cancer, non-Hodgkin’s lymphoma and neoplasms showing resistance to cisplatin and carboplatin [37]. High efficacy in the treatment of advanced and platinum-resistant cancers of the colon has been demonstrated after combining oxaliplatin with 5-fluorouracil and leucovorin [52]. Currently, oxaliplatin is successfully used in the treatment of gastric cancer, however, clinical trials are still being conducted to evaluate the effectiveness of oxaliplatin therapy in combination, among others with S-1 (a prodrug of 5-fluorouracil) [53, 54]. Oxaliplatin, like other platinum drugs, can also cause side effects. Chelation of extracellular Ca2+ ion may disrupt the function of sodium channels and induce acute or chronic peripheral neuropathy [42, 51].

Nedaplatin

Nedaplatin is a cisplatin analogue, a second-generation drug developed in 1983 in Japan. Like cisplatin and carboplatin, its chemical structure has two amine ligands and additionally a glycolic acid dianion [55]. Nedaplatin undergoes hydrolysis, which leads to the formation of a pharmacologically active secondary metabolite (cis-diaminadihydroxyplatin [II]), analogous to cisplatin and carboplatin, capable of forming coordination bonds with DNA bases [43]. This compound is characterized by a reduced nephrotoxic effect and therapeutic effectiveness similar to carboplatin [55]. Nedaplatin has a beneficial effect on squamous cells in lung cancer, head and neck cancers. This drug can be used in patients with recurrent changes of cervical and ovarian cancer after treatment with cisplatin [56]. Nedaplatin can also be used in patients with hypersensitivity reactions to carboplatin therapy [57]. In clinical trials, nedaplatin has been shown to be highly effective pharmacologically in combination with radiotherapy, paclitaxel and irinotecan [58–60]. In combination therapy, its high effectiveness has also been found in the treatment of malignant urological tumors [61].

Lobaplatin

In turn, lobaplatin is a third generation platinum drug. In China, this drug has been approved for the treatment of advanced breast cancer, small-cell lung cancer and chronic myeloid leukemia [62, 63]. According to the literature, lobaplatin is also effective in the treatment of malignant neoplasms of the ovary, cervix, large intestine and stomach [64, 65]. It has been observed that in the treatment of gastric cancer, the antitumor effect of lobaplatin may enhance its combined effect with paclitaxel [62]. It is possible that paclitaxel enhances the effect of lobaplatin and reduces drug resistance by inhibiting the PI3K/Akt pathway also in lung cancer cells [66]. Increased sensitivity of cancer cells to radiation and to the pro-apoptotic activity of lobaplatin was revealed in its cumulative action with radiotherapy [67]. The preliminary results of in vitro and in vivo studies show that lobaplatin may have antitumor efficacy higher than carboplatin, with limited nephro-, neuro- and ototoxicity [68]. Nevertheless, a side effect limiting the use of the drug is the found thrombocytopenia [69].

Heptaplatin

Heptaplatin is a 3rd generation platinum complex with a slight undesirable nephrotoxic effect. It was assumed that the mechanism of its therapeutic action was similar to that of cisplatin and oxaliplatin [70]. In Korea, this drug has been approved for the treatment of advanced stomach tumors [71]. A wide spectrum of anti-tumor activity of heptaplatin was observed in phase I and II clinical trials against gastric, head and neck cancer cells, also in combination therapy with 5-fluorouracil and leucovorin [72–74]. Heptaplatin has also been shown to be effective in the treatment of L1210 cisplatin-resistant leukemia cells [72]. Presumably, the activity of the drug may be partly related to the decreased expression of metallothioneins as a result of heptaplatin action [72].

However, the exact mechanisms of the biological actions of heptaplatin have not yet been fully elucidated.

Therapeutic strategies for new platinum complexes

Since the treatment of neoplastic diseases with classic platinum drugs, apart from their high efficiency, is burdened with many side effects, the search for new platinum complexes, analogous to cisplatin but with low toxicity, is still being sought. Promising platinum compounds at the stage of clinical trials include, among others: picoplatin and phenanthriplatin.

Picoplatin

A new generation of platinum (II) compounds with significant anti-cancer potential is picoplatin. The mechanism of action of picoplatin is similar to that of cisplatin. It consists in creating specific bonds with DNA, although the resulting adducts show greater selectivity of action [37]. The 2-methylpyridine group present in the chemical structure of the compound slows down its intracellular hydrolysis and binding to DNA, which may possibly affect the profile of pharmacological activity and reduced toxicity of the complex [75]. Picoplatin has been shown to be highly effective in treating ovarian and lung cancer that are resistant to cisplatin and carboplatin treatment [76]. This compound was subjected to phase II and III clinical trials, in which the antitumor activity of picoplatin in the treatment of small-cell lung cancer was assessed. In contrast, phase I clinical trials focused on the efficacy of picoplatin in monotherapy in non-haematological malignancies and in combination with 5-fluorouracil and leucovorin in the treatment of colorectal cancer. Phase I studies also focused on the combination of picoplatin and docetaxel in the treatment of hormone refractory prostate cancer and the cumulative effect with liposomal doxorubicin in the treatment of lymphoma and small intestine cancer [77].

Phenanthriplatin

A monofunctional platinum (II) complex, implemented to overcome the mechanisms of cancer cell resistance, is phenanthriplatin, which contains a phenanthridine ligand in its structure. This complex, by means of covalent bonds, with high efficiency, forms adducts with DNA, a result of which means it strongly inhibits the transcription process [19]. Presumably, the DNA-binding profile of phenanthriplatin is different from that of cisplatin, which influences its different biological activity [78]. Although the mechanism of action of phenanthriplatin has not been fully established, it has been observed that this compound may act on cancer cells with greater efficiency than cisplatin and oxaliplatin [79]. The interaction of the complex with organic cation transporters (OCT) contributes to the strong effect of phenanthriplatin on tumor cells, which may suggest that OCT overexpressing tumor cells (e.g. colon cancer) are particularly sensitive to the therapeutic effect of this compound [80]. Moreover, the increased cytotoxic activity of the complex may result from increased cellular uptake [79]. Phenanthriplatin-induced cell death may also result from impaired ribosome biogenesis and increased activation of the L11 ribosomal protein, which, by inhibiting Mdm2 binding to p53, triggers an apoptotic signal [81]. Phenanthriplatin inhibits the mechanisms related to the development of cellular resistance, therefore it may be effective in cancer cells resistant to cisplatin therapy [79, 80]. The beneficial effect of phenanthriplatin has been demonstrated, among others, by on small-cell lung cancer lines [78, 82, 83]. However, in preliminary analyses of clinical trials, significant adverse effects caused by phenanthriplatin were observed, therefore the assessment of its cytotoxic properties is still based on ongoing experimental studies.

Platinum (IV) complexes in anti-cancer therapy

In order to change the biological and chemical properties and improve the pharmacokinetic effects of platinum drugs, the degree of platinum oxidation was modified. In addition to platinum (II) compounds, platinum (IV) compounds have been synthesized. The literature data show that changing the geometry of the molecule from polar to octahedral results in the production of compounds with specific pharmacological properties [15]. Platinum (IV) derivatives are defined as prodrugs which, when reduced to Pt (II) forms, are only activated inside the cell [84]. Platinum (IV) compounds are characterized by increased kinetic activity, lipophilicity and stability, relatively low toxicity and increased activity against drug-resistant cells [85]. The advantage of platinum (IV) complexes is the possibility of their oral use, which can significantly facilitate the form of therapy and improve the quality of life of patients [85].

Satraplatin

Satraplatin is an example of a platinum (IV) complex. It is an analog of carboplatin containing two acetyl groups in its chemical structure, this largely contributes to the increased lipophilicity of the compound and its bioavailability [43]. Increased intracellular biotransformation of satraplatin leads to the formation of the active metabolite (JM118, PtCl2 [NH3] [cha]) [37, 61]. The spectrum of anti-cancer activity of this drug includes platinum-resistant cancer cells of the cervix, prostate, ovary and lungs [61]. The clinical trials performed included the I, II and III phase of the assessment of the antitumor effect in the treatment of prostate cancer, both as monotherapy and in combination with prednisone. In turn, the effect of co-administration of satraplatin with erlotinib and paclitaxel was assessed in relation to breast and lung cancer. Phase I studies also assessed the efficacy of satraplatin in combination with docetaxel and paclitaxel in the treatment of advanced solid tumors. Detailed studies are currently underway to determine the effect of satraplatin in the treatment of patients with high-risk prostate cancer [77].

Adamplatin

An analog of satraplatin with an equally high lipophilicity is adamplatin (IV). This compound is characterized by a broad spectrum of activity, through increased accumulation inside cells, strong inhibition of DNA polymerization and impaired repair of DNA structure damage [15]. Increased cellular uptake of the complex also influences the anti-cancer effect of adamplatin [86]. In addition, sulfur-containing compounds may play a less important role in the mechanisms of cell resistance to adamplatin than to cisplatin. Significant cytotoxic activity of adamplatin was found in in vitro studies against colon cancer cells, leukemia and ovaries, as well as cell lines resistant to cisplatin therapy [84]. Currently, adamplatin has not been implemented in clinical trials.

Oxoplatin

Another platinum (IV) compound with potential anti-cancer properties is oxoplatin. This complex was first synthesized in 1927 by Chugaev and Khlopin [85]. Oxoplatin is activated inside the cell in the presence of ascorbic acid and hydrogen chloride, therefore, oral administration of the compound can significantly accelerate and enhance its biological activity [87]. The increased distribution of this complex in the blood helps to quickly reach the target site of action. The in vitro studies conducted so far show that oxoplatin has a prolonged therapeutic effect and higher pharmacokinetic activity than cisplatin. It has also been found that it inhibits the growth of neoplastic tumors more than cisplatin and may weaken the process of distant metastases [85]. The cytotoxic properties of oxoplatin have been observed in vitro in neoplastic cells of the pancreas, colon, prostate and stomach [85, 88]. However, the mechanism of action of this compound has, as yet, not been fully determined.

Ethacraplatin

So far, the spectrum of the biological and pharmacotherapeutic effects of ethacraplatin (IV) on cancer cells is unexplained. Ethacraplatin is a cisplatin molecule linked to two ethacrynic acid ligands and has the ability to inhibit the activity of glutathione transferase (GST) [89]. This mechanism of action accelerates the formation of adducts with DNA, promotes damage, reduces cellular resistance and thus enhances the pro-apoptotic properties of the complex. However, ethacraplatin is characterized by antitumor activity with a relatively short duration of action [90]. Therefore, work is currently underway to develop encapsulated ethacraplatin, which will increase the amount of the preparation directly in neoplastic tissues, which may contribute to the improvement of therapeutic efficacy in platinum-resistant cells [89].

Liposomal platinum complexes

In order to reduce the nephrotoxicity of platinum complexes and increase their antitumor activity, drug carriers have been designed, e.g. liposomes. To date, two liposomal CDDP compounds have been developed: SPI-077 and lipoplatin [61, 91].

The SPI-077 preparation was characterized by an extended half-life and a fairly good tolerance of the organism, but it showed a relatively low therapeutic activity observed in phase I and II clinical trials. The insufficient therapeutic effect of SPI-
-077 was most likely due to the limited release of cisplatin from liposomes inside the tumors [92].

Lipoplatin

Clinical trials are currently underway to assess the therapeutic properties of lipoplatin. Lipoplatin nanoparticles with a diameter of 110 nm, covered with a polymer coating, consist of a lipid envelope and a central core composed of a cisplatin molecule. The lipid layer consists of: cholesterol, phosphatidylcholine, soybeans, dipalmitoylphosphatidylglycerol and methoxypolyethyleneglycol conjugated with disteroylphosphatidylethanolamine [91, 93, 94]. The lipid layer of nanoparticles facilitates their transport to cancer cells by endocytosis [95]. The mechanism of action of lipoplatin is based on the increased accumulation of cisplatin molecules in the tissues of primary tumors and metastatic sites. On the other hand, the accumulation of the drug in neighbouring healthy cells was many times lower [92, 95]. This is because the drug can penetrate the blood vessel endothelium directly within the neoplastic lesions [93]. By releasing the cisplatin molecule, lipoplatin activates the mitochondrial apoptotic pathway [95]. The liposomal form of cisplatin reduces systemic toxicity and enhances the anti-cancer effect by targeting the drug directly into the tumor [94]. To date, phase I and II clinical trials have been conducted to assess the anti-tumor effect of liposomal cisplatin as monotherapy in the treatment of osteosarcoma and phase III of the treatment of pancreatic cancer in combination with fluorouracil and gemcitabine. The phase I clinical trials also concerned the efficacy of lipoplatin in patients with pleural malignancies after verteporfin therapy. The research evaluating the effects of liposomal cisplatin in the treatment of advanced and refractory solid tumors (phase I) and breast, prostate and skin cancer (phase II) are still ongoing [77].

Multi-core platinum complexes

New therapeutic possibilities are also created by platinum multi-core complexes, which have been developed to increase the platinum-to-DNA binding capacity. Among them, one can distinguish two- and three-core complexes containing at least two platinum atoms in their structure [96]. The BBR3464 gained the greatest recognition among multi-core complexes.

BBR3464

In the chemical structure of the compound, based on the structure of cisplatin, there are two monofunctional groups [trans-PtCl(NH3)2]platinum, linked by platinum tetra-amine (trans-Pt(NH3)2(NH2(CH2)6NH2)2)2+ [97]. The unique structure of BBR3464 ensures increased cellular uptake and enhanced DNA binding resulting from the appropriate electrostatic interaction and the formation of hydrogen bonds [97, 98]. BBR3464 adducts formed from DNA differ significantly from those created as a result of fusion with cisplatin, which may provide a higher therapeutic activity of the complex [98]. This compound is characterized by a prolonged pharmacological effect, which results in the inhibition of the growth of neoplastic tumors even after the end of therapy. This may indicate that the mechanism of action of BBR3464 on cell cycle disorders is different from that of cisplatin [97, 99]. In vitro and in vivo studies have demonstrated the high efficacy of BBR3464 in cisplatin sensitive and resistant tumors, as well as in cells with a mutation of the p53 oncosuppressive gene [99]. Proapoptotic properties of BBR3464 have been observed against ovarian cancer cells and malignant melanoma [97, 100]. In preclinical studies, the cytotoxic activity of BBR3464 was noted at concentrations several times lower than in cisplatin [75]. However, in a clinical evaluation, the antitumor activity of the complex was diversified, depending on the type of neoplastic cells. BBR3464’s inadequate efficacy in some cancers, including gastric cancer may result from the increased metabolic degradation of the compound, which leads to the development of a more appropriate pharmacokinetic profile [97]. To date, BBR3464 has undergone phase II clinical trials to determine its cytotoxic effect against adenocarcinoma of the pancreas and small-cell lung cancer [77].

Conclusions

The discovery and approval of cisplatin for the treatment of cancer at the turn of the 20th century was of great importance for modern oncological medicine. Currently, cisplatin is one of the basic cytostatics used in the treatment of neoplastic diseases, both as monotherapy and in multi-drug therapy in combination with other anticancer compounds. Understanding the broad spectrum of anti-cancer activity of cisplatin has contributed to the search for new platinum complexes showing high therapeutic efficacy, with limited side effects, and the possibility of overcoming platinum resistance. The unique properties of the new platinum complexes may contribute to their wider use in anti-cancer therapy in the future.

Conflict of interest: none declared

Anna Kopacz-Bednarska

Jan Kochanowski University

Institute of Biology

Department of Medical Biology

ul. Uniwersytecka 7

25-406 Kielce, Poland

e-mail: anna.kopacz-bednarska@ujk.edu.pl

Received: 16 Aug 2021
Accepted: 4 Jan 2022

References

  1. Wojciechowska U, Czaderny K, Ciuba A. Cancer in Poland in 2016. Polish National Cancer Registry Department of Epidemiology and Cancer Prevention. 2018: 198.
  2. Didkowska J, Wojciechowska U, Czaderny K. Cancer in Poland in 2017. Polish National Cancer Registry Department of Epidemiology and Cancer Prevention. 2019 : 196.
  3. Strzelecki Z. Prognozy rozwoju chorób nowotworowych w Polsce. In: Potrykowska A, Strzelecki Z, Szymborski J. et al. ed. Zachorowalność i umieralność na nowotwory a sytuacja demograficzna Polski. Zakład Wydawnictw Statystycznych, Warszawa 2014: 717.
  4. Wiszniewska M, Lipinska-Ojrzanowska A, Witkowska A, et al. Occupational cancers Epidemiology and certification. Medycyna Pracy. 2017, doi: 10.13075/mp.5893.00620.
  5. Moreno-Smith M, Lutgendorf SK, Sood AK. Impact of stress on cancer metastasis. Future Oncol. 2010; 6(12): 18631881, doi: 10.2217/fon.10.142, indexed in Pubmed: 21142861.
  6. Soung N, Kim B. Psychological stress and cancer. J Anal Sci Technol. 2015; 6(1), doi: 10.1186/s40543-015-0070-5.
  7. Dai S, Mo Y, Wang Y, et al. Chronic Stress Promotes Cancer Development. Front Oncol. 2020; 10: 1492, doi: 10.3389/fonc.2020.01492, indexed in Pubmed: 32974180.
  8. Key TJ, Schatzkin A, Willett WC, et al. Diet, nutrition and the prevention of cancer. Public Health Nutr. 2004; 7(1A): 187200, doi: 10.1079/phn2003588, indexed in Pubmed: 14972060.
  9. Tandon M, Siddique RA, Singh NK, et al. Anti-cancer diet: reviewing the role of nutrition in cancer prevention. Curr Top Nutraceut Res. 2008; 6(2): 6782.
  10. Nurgali K, Jagoe RT, Abalo R. Editorial: Adverse Effects of Cancer Chemotherapy: Anything New to Improve Tolerance and Reduce Sequelae? Front Pharmacol. 2018; 9: 245, doi: 10.3389/fphar.2018.00245, indexed in Pubmed: 29623040.
  11. Schirrmacher V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review). Int J Oncol. 2019; 54(2): 407419, doi: 10.3892/ijo.2018.4661, indexed in Pubmed: 30570109.
  12. Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019; 2: 141160, doi: 10.20517/cdr.2019.10, indexed in Pubmed: 34322663.
  13. Bukowski K, Kciuk M, Kontek R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int J Mol Sci. 2020; 21(9), doi: 10.3390/ijms21093233, indexed in Pubmed: 32370233.
  14. Dai Z, Wang Z. Photoactivatable Platinum-Based Anticancer Drugs: Mode of Photoactivation and Mechanism of Action. Molecules. 2020; 25(21), doi: 10.3390/molecules25215167, indexed in Pubmed: 33171980.
  15. Śliwińska-Hill U, Szumełda M. Biologiczne podstawy terapii przeciwnowotworowej z zastosowaniem leków platynowych. Oddziaływanie z cytochromem c. Nowotwory. Journal of Oncology. 2016; 66(2): 136150, doi: 10.5603/njo.2016.0023.
  16. Makovec T. Cisplatin and beyond: molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol Oncol. 2019; 53(2): 148158, doi: 10.2478/raon-2019-0018, indexed in Pubmed: 30956230.
  17. Florea AM, Büsselberg D. Cisplatin as an anti-tumor drug: cellular mechanisms of activity, drug resistance and induced side effects. Cancers (Basel). 2011; 3(1): 13511371, doi: 10.3390/cancers3011351, indexed in Pubmed: 24212665.
  18. Tchounwou PB, Dasari S, Noubissi FK, et al. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014; 740: 364378, doi: 10.1016/j.ejphar.2014.07.025, indexed in Pubmed: 25058905.
  19. Gregory MT, Park GaY, Johnstone TC, et al. Understanding and improving platinum anticancer drugs--phenanthriplatin. Anticancer Res. 2014; 34(1): 471476, indexed in Pubmed: 24403503.
  20. Velma V, Dasari SR, Tchounwou PB. Low Doses of Cisplatin Induce Gene Alterations, Cell Cycle Arrest, and Apoptosis in Human Promyelocytic Leukemia Cells. Biomark Insights. 2016; 11: 113121, doi: 10.4137/BMI.S39445, indexed in Pubmed: 27594783.
  21. Konac E, Varol N, Kiliccioglu I, et al. Synergistic effects of cisplatin and proteasome inhibitor bortezomib on human bladder cancer cells. Oncol Lett. 2015; 10(1): 560564, doi: 10.3892/ol.2015.3250, indexed in Pubmed: 26171069.
  22. Perkhofer L, Berger AW, Beutel AK, et al. Nal-IRI with 5-fluorouracil (5-FU) and leucovorin or gemcitabine plus cisplatin in advanced biliary tract cancer - the NIFE trial (AIO-YMO HEP-0315) an open label, non-comparative, randomized, multicenter phase II study. BMC Cancer. 2019; 19(1): 990, doi: 10.1186/s12885-019-6142-y, indexed in Pubmed: 31646981.
  23. Yeku O, Russo AL, Lee H, et al. A phase 2 study of combined chemo-immunotherapy with cisplatin-pembrolizumab and radiation for unresectable vulvar squamous cell carcinoma. J Transl Med. 2020; 18(1): 350, doi: 10.1186/s12967-020-02523-5, indexed in Pubmed: 32928237.
  24. Ciarimboli G, Ciarimboli G. Membrane transporters as mediators of Cisplatin effects and side effects. Scientifica (Cairo). 2012; 2012(1): 473829550, doi: 10.6064/2012/473829, indexed in Pubmed: 24278698.
  25. Łacko A, Hudziec P, Mazur G. Porównanie parametrów farmakologicznych i klinicznych cisplatyny i karboplatyny w leczeniu guzów litych. Nowotwory J Oncol. 2000; 50(6): 609.
  26. Aldossary S. Review on Pharmacology of Cisplatin: Clinical Use, Toxicity and Mechanism of Resistance of Cisplatin. Biomed Pharmacol J. 2019; 12(1): 0715, doi: 10.13005/bpj/1608.
  27. Pabla N, Dong Z. Cisplatin nephrotoxicity: mechanisms and renoprotective strategies. Kidney Int. 2008; 73(9): 9941007, doi: 10.1038/sj.ki.5002786, indexed in Pubmed: 18272962.
  28. Volarevic V, Djokovic B, Jankovic MG, et al. Molecular mechanisms of cisplatin-induced nephrotoxicity: a balance on the knife edge between renoprotection and tumor toxicity. J Biomed Sci. 2019; 26(1): 25, doi: 10.1186/s12929-019-0518-9, indexed in Pubmed: 30866950.
  29. Park MS, De Leon M, Devarajan P. Cisplatin induces apoptosis in LLC-PK1 cells via activation of mitochondrial pathways. J Am Soc Nephrol. 2002; 13(4): 858865, doi: 10.1681/ASN.V134858, indexed in Pubmed: 11912244.
  30. Jiang M, Yi X, Hsu S, et al. Role of p53 in cisplatin-induced tubular cell apoptosis: dependence on p53 transcriptional activity. Am J Physiol Renal Physiol. 2004; 287(6): F1140F1147, doi: 10.1152/ajprenal.00262.2004, indexed in Pubmed: 15315938.
  31. Rathinam R, Ghosh S, Neumann WL, et al. Cisplatin-induced apoptosis in auditory, renal, and neuronal cells is associated with nitration and downregulation of LMO4. Cell Death Discov. 2015; 1, doi: 10.1038/cddiscovery.2015.52, indexed in Pubmed: 26925255.
  32. Wang P, Cui J, Wen J, et al. Cisplatin induces HepG2 cell cycle arrest through targeting specific long noncoding RNAs and the p53 signaling pathway. Oncol Lett. 2016; 12(6): 46054612, doi: 10.3892/ol.2016.5288, indexed in Pubmed: 28105167.
  33. Galluzzi L, Senovilla L, Vitale I, et al. Molecular mechanisms of cisplatin resistance. Oncogene. 2012; 31(15): 18691883, doi: 10.1038/onc.2011.384, indexed in Pubmed: 21892204.
  34. Sarin N, Engel F, Kalayda GV, et al. Cisplatin resistance in non-small cell lung cancer cells is associated with an abrogation of cisplatin-induced G2/M cell cycle arrest. PLoS One. 2017; 12(7): e0181081, doi: 10.1371/journal.pone.0181081, indexed in Pubmed: 28746345.
  35. Bahar E, Kim JY, Kim HS, et al. Establishment of Acquired Cisplatin Resistance in Ovarian Cancer Cell Lines Characterized by Enriched Metastatic Properties with Increased Twist Expression. Int J Mol Sci. 2020; 21(20), doi: 10.3390/ijms21207613, indexed in Pubmed: 33076245.
  36. Wheate NJ, Walker S, Craig GE, et al. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalton Trans. 2010; 39(35): 81138127, doi: 10.1039/c0dt00292e, indexed in Pubmed: 20593091.
  37. Subocz M, Popławska B, Bielawska A, et al. Pochodne platyny w chemioterapii chorób nowotworowych. Annal Acad Med Siles. 2011; 65(4): 7076.
  38. Ciancetta A, Coletti C, Marrone A, et al. Activation of carboplatin by chloride ions: a theoretical investigation. Theor Chem Acc. 2011; 129(6): 757769, doi: 10.1007/s00214-011-0933-9.
  39. Sousa G, Wlodarczyk S, Monteiro G. Carboplatin: molecular mechanisms of action associated with chemoresistance. Braz J Pharm Sci. 2014; 50(4): 693701, doi: 10.1590/s1984-82502014000400004.
  40. Pavelka M, Lucas MF, Russo N. On the hydrolysis mechanism of the second-generation anticancer drug carboplatin. Chemistry. 2007; 13(36): 1010810116, doi: 10.1002/chem.200700887, indexed in Pubmed: 17896336.
  41. Werengowska KM, Wiśniewski M, Terzyk AP, et al. Chemiczne aspekty celowanej terapii przeciwnowotworowej II. Połączenia nośnik-lek. Wiad Chem. 2012; 66(7-8): 637670.
  42. Kanat O, Ertas H, Caner B. Platinum-induced neurotoxicity: A review of possible mechanisms. World J Clin Oncol. 2017; 8(4): 329335, doi: 10.5306/wjco.v8.i4.329, indexed in Pubmed: 28848699.
  43. Trynda-Lemiesz L, Śliwińska-Hill U. Kompleksy metali w terapii nowotworowej. Teraźniejszość i przyszłość. Nowotwory J Oncol. 2011; 61(5): 465474.
  44. Huang CY, Cheng M, Lee NR, et al. Comparing Paclitaxel-Carboplatin with Paclitaxel-Cisplatin as the Front-Line Chemotherapy for Patients with FIGO IIIC Serous-Type Tubo-Ovarian Cancer. Int J Environ Res Public Health. 2020; 17(7), doi: 10.3390/ijerph17072213, indexed in Pubmed: 32224896.
  45. Rosca L, Robert-Boire V, Delisle JF, et al. Carboplatin and vincristine neurotoxicity in the treatment of pediatric low-grade gliomas. Pediatr Blood Cancer. 2018; 65(11): e27351, doi: 10.1002/pbc.27351, indexed in Pubmed: 30014595.
  46. Yu KD, Ye FG, He M, et al. Effect of Adjuvant Paclitaxel and Carboplatin on Survival in Women With Triple-Negative Breast Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020; 6(9): 13901396, doi: 10.1001/jamaoncol.2020.2965, indexed in Pubmed: 32789480.
  47. Mielcarek P, Majdak E, Krasińska E, et al. Comparison of quality of life in patients with advanced ovarian cancer treated with intravenous paclitaxel and carboplatin versus cyclophosphamide and cisplatin as first line chemotherapy a preliminary report. Nowotwory J Oncol. 2002; 52(1): 3336.
  48. Alcindor T, Beauger N. Oxaliplatin: a review in the era of molecularly targeted therapy. Curr Oncol. 2011; 18(1): 1825, doi: 10.3747/co.v18i1.708, indexed in Pubmed: 21331278.
  49. Kozakiewicz K, Kaczmarczyk M. Cisplatyna lek z przypadku. Curr Gynecol Oncol. 2012; 10(2): 131140.
  50. Arango D, Wilson AJ, Shi Q, et al. Molecular mechanisms of action and prediction of response to oxaliplatin in colorectal cancer cells. Br J Cancer. 2004; 91(11): 19311946, doi: 10.1038/sj.bjc.6602215, indexed in Pubmed: 15545975.
  51. Seetharam Rn, Sood A, Goel S. Oxaliplatin: pre-clinical perspectives on the mechanisms of action, response and resistance. Ecancermedicalscience. 2009; 3: 153, doi: 10.3332/ecancer.2009.153, indexed in Pubmed: 22276017.
  52. Tournigand C, André T, Bonnetain F, et al. Multicenter International Study of Oxaliplatin/5-Fluorouracil/Leucovorin in the Adjuvant Treatment of Colon Cancer (MOSAIC) Investigators. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 2004; 350(23): 23432351, doi: 10.1056/NEJMoa032709, indexed in Pubmed: 15175436.
  53. Saeki H, Emi Y, Oki E, et al. Kyushu Study group of Clinical Cancer (KSCC). Study protocol of a phase II clinical trial (KSCC1501A) examining oxaliplatin + S-1 for treatment of HER2-negative advanced/recurrent gastric cancer previously untreated with chemotherapy. BMC Cancer. 2018; 18(1): 57, doi: 10.1186/s12885-017-3937-6, indexed in Pubmed: 29310611.
  54. Xiao C, Qian J, Zheng Y, et al. A phase II study of biweekly oxaliplatin plus S-1 combination chemotherapy as a first-line treatment for patients with metastatic or advanced gastric cancer in China. Medicine (Baltimore). 2019; 98(20): e15696, doi: 10.1097/MD.0000000000015696, indexed in Pubmed: 31096513.
  55. Shimada M, Itamochi H, Kigawa J. Nedaplatin: a cisplatin derivative in cancer chemotherapy. Cancer Manag Res. 2013; 5: 6776, doi: 10.2147/CMAR.S35785, indexed in Pubmed: 23696716.
  56. Mabuchi S, Kimura T. Nedaplatin: a radiosensitizing agent for patients with cervical cancer. Chemother Res Pract. 2011; 2011: 963159, doi: 10.1155/2011/963159, indexed in Pubmed: 22312560.
  57. Chikazawa K, Netsu S, Imai K, et al. Nedaplatin use in patients with hypersensitivity reaction episodes to carboplatin. Taiwan J Obstet Gynecol. 2020; 59(4): 546550, doi: 10.1016/j.tjog.2020.05.013, indexed in Pubmed: 32653127.
  58. Kenmotsu Y, Oshita F, Sugiura M, et al. Nedaplatin and irinotecan in patients with large-cell neuroendocrine carcinoma of the lung. Anticancer Res. 2012; 32(4): 14531456, indexed in Pubmed: 22493385.
  59. Ge Li, Li N, Yuan GW, et al. Nedaplatin and paclitaxel compared with carboplatin and paclitaxel for patients with platinum-sensitive recurrent ovarian cancer. Am J Cancer Res. 2018; 8(6): 10741082, indexed in Pubmed: 30034944.
  60. Zhu H, Ge X, Lu Y, et al. Nedaplatin-based chemotherapy regimens combined with concurrent radiotherapy as first-line treatment for stage II-III esophageal squamous cell carcinoma. Oncol Lett. 2019; 17(1): 594602, doi: 10.3892/ol.2018.9564, indexed in Pubmed: 30655806.
  61. Ndagi U, Mhlongo N, Soliman ME. Metal complexes in cancer therapy - an update from drug design perspective. Drug Des Devel Ther. 2017; 11: 599616, doi: 10.2147/DDDT.S119488, indexed in Pubmed: 28424538.
  62. Hua S, Kong X, Chen B, et al. Anticancer Mechanism of Lobaplatin as Monotherapy and in Combination with Paclitaxel in Human Gastric Cancer. Curr Mol Pharmacol. 2018; 11(4): 316325, doi: 10.2174/1874467211666180813095050, indexed in Pubmed: 30101722.
  63. Zhou NN, Zhao YY, Zhai LZ, et al. The Efficacy and Toxicity of Lobaplatin-contained Chemotherapy in Extensive-stage Small-cell Lung Cancer. J Cancer. 2018; 9(13): 22322236, doi: 10.7150/jca.24557, indexed in Pubmed: 30026818.
  64. Yin CY, Lin XL, Tian L, et al. Lobaplatin inhibits growth of gastric cancer cells by inducing apoptosis. World J Gastroenterol. 2014; 20(46): 1742617433, doi: 10.3748/wjg.v20.i46.17426, indexed in Pubmed: 25516654.
  65. Shan L, Bai B, Lv Y, et al. Lobaplatin suppresses proliferation and peritoneal metastasis of colorectal cancer in a preclinical model. Biomed Pharmacother. 2018; 108: 486491, doi: 10.1016/j.biopha.2018.09.063, indexed in Pubmed: 30243080.
  66. Ma D, Li S, Cui Y, et al. Paclitaxel increases the sensitivity of lung cancer cells to lobaplatin via PI3K/Akt pathway. Oncol Lett. 2018; 15(5): 62116216, doi: 10.3892/ol.2018.8086, indexed in Pubmed: 29616103.
  67. Pan S, Sun Y, Sui D, et al. Lobaplatin promotes radiosensitivity, induces apoptosis, attenuates cancer stemness and inhibits proliferation through PI3K/AKT pathway in esophageal squamous cell carcinoma. Biomed Pharmacother. 2018; 102: 567574, doi: 10.1016/j.biopha.2018.03.109, indexed in Pubmed: 29597090.
  68. Zhou Z, Jiang H, Xia J, et al. Comparison of the therapeutic effects of lobaplatin and carboplatin on retinoblastoma in vitro and in vivo. Int J Oncol. 2020; 57(3): 697706, doi: 10.3892/ijo.2020.5085, indexed in Pubmed: 32582992.
  69. Welink J, Boven E, Vermorken JB, et al. Pharmacokinetics and pharmacodynamics of lobaplatin (D-19466) in patients with advanced solid tumors, including patients with impaired renal of liver function. Clin Cancer Res. 1999; 5(9): 23492358, indexed in Pubmed: 10499604.
  70. Zhang HY, Liu YR, Li W, et al. Condensations of single DNA molecules induced by heptaplatin and its chiral isomer. AIP Advances. 2014; 4(8): 087128, doi: 10.1063/1.4893672.
  71. Kang X, Xiao HH, Song HQ, et al. Advances in drug delivery system for platinum agents based combination therapy. Cancer Biol Med. 2015; 12(4): 362374, doi: 10.7497/j.issn.2095-3941.2015.0063, indexed in Pubmed: 26779373.
  72. Choi CH, Cha YJ, An CS, et al. Molecular mechanisms of heptaplatin effective against cisplatin-resistant cancer cell lines: less involvement of metallothionein. Cancer Cell Int. 2004; 4(1): 6, doi: 10.1186/1475-2867-4-6, indexed in Pubmed: 15494073.
  73. Lee WS, Lee GW, Kim HW, et al. A phase II trial of haptaplatin/5-FU and leucovorin for advanced stomach cancer. Cancer Res Treat. 2005; 37(4): 208211, doi: 10.4143/crt.2005.37.4.208, indexed in Pubmed: 19956515.
  74. Lee KH, Hyun MS, Kim HK, et al. Randomized, multicenter, phase III trial of heptaplatin 1-hour infusion and 5-fluorouracil combination chemotherapy comparing with cisplatin and 5-fluorouracil combination chemotherapy in patients with advanced gastric cancer. Cancer Res Treat. 2009; 41(1): 1218, doi: 10.4143/crt.2009.41.1.12, indexed in Pubmed: 19688066.
  75. Brown SD, Trotter KD, Sutcliffe OB, et al. Combining aspects of the platinum anticancer drugs picoplatin and BBR3464 to synthesize a new family of sterically hindered dinuclear complexes; their synthesis, binding kinetics and cytotoxicity. Dalton Trans. 2012; 41(37): 1133011339, doi: 10.1039/c2dt31313h, indexed in Pubmed: 22886151.
  76. Tang CH, Parham C, Shocron E, et al. Picoplatin overcomes resistance to cell toxicity in small-cell lung cancer cells previously treated with cisplatin and carboplatin. Cancer Chemother Pharmacol. 2011; 67(6): 13891400, doi: 10.1007/s00280-010-1435-5, indexed in Pubmed: 20809122.
  77. www.clinicaltrials.gov.
  78. Monroe JD, Hruska HL, Ruggles HK, et al. Anti-cancer characteristics and ototoxicity of platinum(II) amine complexes with only one leaving ligand. PLoS One. 2018; 13(3): e0192505, doi: 10.1371/journal.pone.0192505, indexed in Pubmed: 29513752.
  79. Park GaY, Wilson JJ, Song Y, et al. Phenanthriplatin, a monofunctional DNA-binding platinum anticancer drug candidate with unusual potency and cellular activity profile. Proc Natl Acad Sci U S A. 2012; 109(30): 1198711992, doi: 10.1073/pnas.1207670109, indexed in Pubmed: 22773807.
  80. Hucke A, Park GaY, Bauer OB, et al. Interaction of the New Monofunctional Anticancer Agent Phenanthriplatin With Transporters for Organic Cations. Front Chem. 2018; 6: 180, doi: 10.3389/fchem.2018.00180, indexed in Pubmed: 29888219.
  81. Facchetti G, Rimoldi I. Anticancer platinum(II) complexes bearing N-heterocycle rings. Bioorg Med Chem Lett. 2019; 29(11): 12571263, doi: 10.1016/j.bmcl.2019.03.045, indexed in Pubmed: 30935797.
  82. McDevitt CE, Yglesias MV, Mroz AM, et al. Monofunctional platinum(II) compounds and nucleolar stress: is phenanthriplatin unique? J Biol Inorg Chem. 2019; 24(6): 899908, doi: 10.1007/s00775-019-01707-9, indexed in Pubmed: 31494760.
  83. Monroe JD, Moolani SA, Irihamye EN, et al. RNA-Seq Analysis of Cisplatin and the Monofunctional Platinum(II) Complex, Phenanthriplatin, in A549 Non-Small Cell Lung Cancer and IMR90 Lung Fibroblast Cell Lines. Cells. 2020; 9(12), doi: 10.3390/cells9122637, indexed in Pubmed: 33302475.
  84. Kaspárková J, Nováková O, Vrána O, et al. Molecular aspects of antitumor effects of a new platinum(IV) drug. Mol Pharmacol. 2006; 70(5): 17081719, doi: 10.1124/mol.106.027730, indexed in Pubmed: 16896071.
  85. Olszewski U, Ach F, Ulsperger E, et al. In Vitro Evaluation of Oxoplatin: An Oral Platinum(IV) Anticancer Agent. Met Based Drugs. 2009; 2009: 348916, doi: 10.1155/2009/348916, indexed in Pubmed: 19587824.
  86. Halámiková A, Heringová P, Kaspárková J, et al. Cytotoxicity, mutagenicity, cellular uptake, DNA and glutathione interactions of lipophilic trans-platinum complexes tethered to 1-adamantylamine. J Inorg Biochem. 2008; 102(56): 10771089, doi: 10.1016/j.jinorgbio.2007.12.015, indexed in Pubmed: 18237783.
  87. Cerón-Carrasco JP. Theoretical Prediction of Dual-Potency Anti-Tumor Agents: Combination of Oxoplatin with Other FDA-Approved Oncology Drugs. Int J Mol Sci. 2020; 21(13), doi: 10.3390/ijms21134741, indexed in Pubmed: 32635199.
  88. Klameth L, Rath B, Hamilton G. In vitro Cytotoxic Activities of the Oral Platinum(IV) Prodrug Oxoplatin and HSP90 Inhibitor Ganetespib against a Panel of Gastric Cancer Cell Lines. J Cancer. 2017; 8(10): 17331743, doi: 10.7150/jca.17816, indexed in Pubmed: 28819369.
  89. Allocati N, Masulli M, Di Ilio C, et al. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis. 2018; 7(1): 8, doi: 10.1038/s41389-017-0025-3, indexed in Pubmed: 29362397.
  90. Li S, Li C, Jin S, et al. Overcoming resistance to cisplatin by inhibition of glutathione S-transferases (GSTs) with ethacraplatin micelles in vitro and in vivo. Biomaterials. 2017; 144: 119129, doi: 10.1016/j.biomaterials.2017.08.021, indexed in Pubmed: 28834763.
  91. Liu D, He C, Wang AZ, et al. Application of liposomal technologies for delivery of platinum analogs in oncology. Int J Nanomedicine. 2013; 8: 33093319, doi: 10.2147/IJN.S38354, indexed in Pubmed: 24023517.
  92. Zisman N, Dos Santos N, Johnstone S, et al. Optimizing Liposomal Cisplatin Efficacy through Membrane Composition Manipulations. Chemother Res Pract. 2011; 2011: 213848, doi: 10.1155/2011/213848, indexed in Pubmed: 22312548.
  93. Jehn CF, Boulikas T, Kourvetaris A, et al. Pharmacokinetics of liposomal cisplatin (lipoplatin) in combination with 5-FU in patients with advanced head and neck cancer: first results of a phase III study. Anticancer Res. 2007; 27(1A): 471475, indexed in Pubmed: 17352269.
  94. Fantini M, Gianni L, Santelmo C, et al. Lipoplatin Treatment in Lung and Breast Cancer. Chemother Res Pract. 2011; 2011: 17, doi: 10.1155/2011/125192.
  95. Stathopoulos GP, Boulikas T. Lipoplatin formulation review article. J Drug Deliv. 2012; 2012: 581363, doi: 10.1155/2012/581363, indexed in Pubmed: 21904682.
  96. Weiss-Gradzińska W, Krzempek W, Trynda-Lemiesz L. Mechanizm oporności na leki platynowe oraz strategie pokonywania tego zjawiska. Wiad Chem. 2013; 67: 1112.
  97. Manzotti C, Pratesi G, Menta E, et al. BBR 3464: a novel triplatinum complex, exhibiting a preclinical profile of antitumor efficacy different from cisplatin. Clin Cancer Res. 2000; 6(7): 26262634, indexed in Pubmed: 10914703.
  98. Salerno D, Beretta GL, Zanchetta G, et al. Platinum-Based Drugs and DNA Interactions Studied by Single-Molecule and Bulk Measurements. Biophys J. 2016; 110(10): 21512161, doi: 10.1016/j.bpj.2016.02.030, indexed in Pubmed: 27224480.
  99. Sessa C, Capri G, Gianni L, et al. Clinical and pharmacological phase I study with accelerated titration design of a daily times five schedule of BBR3464, a novel cationic triplatinum complex. Ann Oncol. 2000; 11(8): 977983, doi: 10.1023/a:1008302309734, indexed in Pubmed: 11038034.
  100. Kasparkova J, Zehnulova J, Farrell N, et al. DNA Interstrand Cross-links of the Novel Antitumor Trinuclear Platinum Complex BBR3464. J Biol Chem. 2002; 277(50): 4807648086, doi: 10.1074/jbc.m208016200.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Wydawcą serwisu jest VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail: viamedica@viamedica.pl