Open access peer-reviewed chapter

Perspective Chapter: Topoisomerase 1 and Colo Rectal Carcinoma

Written By

Ahmed Mohamed Nabil Helaly and Doaa Ghorab

Submitted: 19 October 2022 Reviewed: 10 November 2022 Published: 22 December 2022

DOI: 10.5772/intechopen.108988

From the Edited Volume

DNA Replication - Epigenetic Mechanisms and Gene Therapy Applications

Edited by Ziyad S. Haidar

Chapter metrics overview

134 Chapter Downloads

View Full Metrics

Abstract

Topoisomerase 1 is the main enzyme playing an important role in relaxing. The supercoiled DNA strands allow the replication fork to transcribe the DNA to RNA and finally control protein production in active and replicating cells. Blocking this essential machinery is a cornerstone mechanism in treating tumors, such as liver, breast, and metastatic colorectal carcinoma. Irinotecan is a topoisomerase inhibitor that blocks the replication ending in DNA break and tumor cell death. This chemotherapy has been successfully used in combination to overcome metastatic colorectal carcinoma. The topoisomerase-1 inhibitor makes a protein DNA complex stuck with the replicating fork creating a single DNA break, unlike topoisomerase-2, which is responsible for double DNA break. This inhibitor is exposed to drug resistance with complex machinery. Drug resistance can occur as a result of altered DNA methylation, changes in topoisomerase expression, histone recombination, or drug export pump. High expression of topoisomerase-1 is a marker of the number of tumors suggesting multiple roles of topoisomerase-1.

Keywords

  • topoisomerase-1
  • irinotecan
  • colorectal carcinoma
  • epigenetic
  • drug resistance
  • supercoiled DNA

1. Introduction

DNA topoisomerases are important enzymes that modify the double-stranded DNA topology. They act in both directions to relax supercoiled DNA and enforce relaxed DNA to be tenser. These 3D structure modifications share in the control of DNA transcription and finally protein translation. In humans, there are two types of isomerases: the first one is topoisomerase-1 (TOP-1); the second one is topoisomerase-2 (TOP-2). Because of the canonical functions of TOPs, these enzymes have been a target for treating overactive cells, such as cancer cells or bacteria that contain different types of isomerases [1]. TOPs covalently bind DNA and create DNA adducts aiming to release the over-twisted DNA strand. Topoisomerase inhibitors (TOP inhibitors) create TOP/DNA obstacles in front of the active replicating fork. The smash between the fork and the DNA protein complex ends in a DNA break. TOP-1 differs from TOP-2 in that the first one is responsible for a single DNA break, while the latter is involved in double DNA damage [2]. In this chapter, we are going to discuss the relationship between TOP-1 and cancer. The chapter aims to put a spot on the role of TOP-1 in cancer colon, and TOP-1 inhibitors’ role to overcome cancer colon. It is important to discuss TOP-1 resistance.

1.1 Topoisomerase-1 and cancer

Since 1989, scientists recorded the high expression of TOP-1 in aggressive tumors such as late-stage colorectal carcinoma. It seems that such tumors use TOP-1 to support their replication machine. These findings were reported in renal tumors or brain neoplasms. The excess TOP-1 activity was highest seen in leukemia besides cancer colon. Moreover, TOP-1 overactivity correlated with poor prognosis in these tumors [3, 4].

TOP-1 may exert complex relation to the process of carcinogenesis. Experimental work demonstrated that knock-out TOP-1 plays a role in oncogenic property. The knock-out cells showed less aging and more replication capacity. Interestingly, the genome was more resistant to DNA damage [5]. More recent work showed opposite results concluding that oxidative stress induces cell aging with excess TOP-1 activity [6]. It is suggested that both senescence and active replication are parts of the process of neoplasia. An immunohistochemical study of secondary pterygium showed both overexpression of TOP-1 and glutathione in the same pathology [7].

A recent study on cancer liver samples using both immunohistochemistry and microarray demonstrated upregulation of TOP-1 and TOP-2 expression in poor prognosis liver cancer candidates. Both genes were involved in the function of the guardian p53 pathway and apoptosis cascade. The study showed that it is a wise idea to consider topoisomerases as oncogenes. Furthermore, these 2 targets are potentially good potentials for cancer chemotherapy [8]. TOP-1 inhibitors include etoposide, camptothecin, and Adriamycin with wide use in clinical practice. More research on different extracts to decrease the side effects and improve the efficacy is going on [9, 10]. Studies in yeast demonstrated that active or aberrant TOP-1 induces DNA mutagenicity, and later on, unstable DNA contributes to cancer development. This mechanism is controlled in mammalians by safety SUMOylation post transcription mechanism [11]. The small ubiquitin-like modifier (SUMO) is a pathway that contributes to transcription, immunity, signaling, and stabilization of the genome. Unfortunately, defective TOP-1 and its regulatory SUMOlyation may end in tumor genesis [12].

Topoisomerase-1 gene expression is considered a marker of drug response in metastatic colorectal carcinoma. It is recommended to use irinotecan (TOP-1 inhibitor) to treat these advanced colorectal tumors where TOP-1 expression is over-expressed. It seems that these tumors over-express TOP-1 to support the tumor DNA repair making the tumor cell resistant to death [13]. TOP-1 inhibitor irinotecan has been tested on cell lines to induce acetylation of the p53 and interrupt the histone deacetylase activity, making the resistant genome of growing cancer cells suitable for DNA break. It seems that TOP-1 inhibition is challenging in the understanding of the cancer colon pathway [14]. Metabolomic analysis of colorectal carcinoma cells treated by irinotecan showed shifting to glycolysis and an increase in oxygen consumption as markers of good response to cancer chemotherapy [15].

1.2 Topoisomerase inhibitors

The TOP-1 inhibitors were first discovered from a tree growing in China named Camptotheca acuminate. The extraction product of this tree was a component of Chinese medicine. Later on, in the 70s and the 80s, research work managed to construct TOP-1 inhibitors in the lab and formulate them as chemotherapy [16].

Drugs inhibiting TOP-1 have been used for decades to treat malignant tumors. These chemotherapies are called TOP-1 poisons. The first-generation candidate of these compounds is camptothecin. Newer generations, include irinotecan, topotecan, and belotecan [17]. Like any chemotherapy, camptothecin derivatives have many side effects. Scientists are working with non-camptothecin generations with fewer hazards [18]. TOP-1 poisons have been prescribed for multiple tumors including colorectal cancer, ovarian tumors, small cell lung malignancy, and myeloid proliferative disorders [19]. Aggressive neoplasms over expressing TOP-1 are suggested to be good candidates for TOP-1 inhibitors as in breast or ovarian cancers besides colorectal malignancy. It is expected that TOP-1 will be radical chemotherapy for these tumors exposing their DNA to break [20, 21, 22]. The researchers were working with TOP-1 inhibitors for half a century. They managed to get the crystal structure of the enzyme. The advances in molecular docking in the last 20 years give scientists a great opportunity to design lots of derivatives to discover new drugs that are more potent with more specific functions aiming to reduce the side effects. However, the difficulty was that the mechanism of action is canonical that it is extremely difficult to avoid hazards. To overcome such obstacle, the advances in drug delivery will reduce the side effects by loading the chemotherapy dose directly onto tumor cells [23].

1.3 Topoisomerase-1 inhibitors and cancer colon

Irinotecan is considered the first drug of choice in treating metastatic colorectal carcinoma in combination with 5 fluorouracil and folinic acid. Irinotecan is a working TOP-1 inhibitor that is metabolized in the liver to a more active compound SN-38. This chemotherapy is characterized by a high volume of distribution. Fortunately, cancer cells have excess carboxylesterase enzymes that can metabolize irinotecan into its active component [24, 25, 26]. Irinotecan is a derivative of camptothecin and was approved for cancer colon in 1994. Its major side effects are neutropenia and diarrhea, which are responsible for dysbiosis. This chemotherapy was approved for children and adults. It is successful for metastatic colorectal neoplasia, as well as, solid tumors. The drug was constructed by the Japanese Yakult Honsha company. The newer generation products have been tested as new chemotherapy but failed in the late clinical phases such as rubitecan, gimatecan, lurtotecan, diflomotecan, elomotecan, silatecan, exatecan, namitecan [27, 28].

New compounds are being verified for TOP-1 inhibitors with better profiles. These compounds include belotecan and gimatecan. The first one is hydrophilic while the latter is dissolved in fat [29, 30].

Belotecan has been used to treat resistant ovarian tumors with better side effects in combination with other new modalities [31]. Other trials have been applied to treat lung carcinoma and biliary tumors [32, 33]. There is little data about the efficacy of belotecan on colorectal neoplasms. On the other hand, gimatecan showed promising results in brain tumors [34].

The advances in nanotechnology and drug delivery can overcome the side effects of traditional TOP-1. Nano liposomal irinotecan has been applied in a pancreatic neoplasm with the hope to achieve success. The results expressed better side effects, but the overall survival represented a weak response [35]. Nano liposomal irinotecan, in combination with other classic chemotherapy, showed better cancer pancreas response. Diarrhea seems to be less counted, but the patients still suffered from neutropenia in a fifth of cases treated to the new model [36].

In regard to colorectal cancer, there are promising clinical trials that liposomal TOP-1 will help in overcoming late-stage conditions. The experimental trials in mice showed better survival rates with fewer side effects [37]. Another strategy is to use a lipophilic active gradient of irinotecan SN38 to suppress advanced colorectal tumors. The results showed a successful modality and the FDA approved the drug for the treatment of late-stage cancer colon [38].

New drugs, which are non-camptothecin derivatives, have been introduced to manage different tumors. These modalities include dibenzonaphthyridines, as well as, indeno isoquinolines. They are more strong stable DNA-protein complexes. Trials of metals such as platinum, gold, copper, zinc, and others have been suggested as TOP-1 modulators [39].

More research work is concerned with indenoisoquinolines, regards the capability to inhibit TOP-1. These new groups of the drug are subjected to structural modification. They represent promising drugs with potentially fewer side effects. It is proposed that the new chemical will have a multi-mechanism of action like nuclear receptors targeting, TOP-2 interaction, modulating estrogen receptors, and manipulating VEGFR and HIF-1 alpha [40]. Studies suggested that indenoisoquinolines are weak TOP-1 inhibitors in combination with other functions. It seems that weak TOP-1 inhibitors in conjunction with other drugs, or even other pathways, are good rationales for modern cancer therapy [41].

1.3.1 Indenoisoquinolines

More than 20 years ago, a group of scientists managed to create a new anti-TOP-1 chemical 6,11-dimethyl-6,11-dihydro-5H-indeno[1,2-c]isoquinolin-5-one. This compounds the parent of quinolone family, and it exerted promising results on different human cell lines [42]. Since that time, dozens of derivatives have been introduced to be tested experimentally. These compounds have the advantage of being flat ones that strongly bind the TOP-1/DNA hybrid. The new compounds bind the TOP-1 at the arginine 364 of the enzyme TOP-1 [43]. These potential new drugs showed stronger cytotoxicity relative to their anti-TOP-1 mechanism and are expected with combined dynamic to have a less toxic profile [44]. Other pathways affected by indenoisoquinolines include induction of cell cycle arrest, stimulation of apoptotic response, and modulation of MAPK cascade. Other potential role includes phosphorylation of JUNK pathway and inhibition of the oncogene MYC pathway [45]. On the other hand, despite the strong multi-dynamic function of these drugs, they did not replace the classic TOP-1. Research is still going to tune drugs with a balance between efficacy and hazard effects.

1.4 Topoisomerase-1 inhibitor resistance

Topoisomerase inhibitors, like other chemotherapy, are subjected to resistance. Many mechanisms are involved in this process. The tumor uses efflux mechanisms to reduce the level of the drug in the tumor mass. Mutations of the TOP-1 make the drug less effective in inhibiting the enzyme. Other strategies include enhancing the DNA repair to overcome TOP-1 poison. The tumor cells stimulate p53 to support the malignant mass to survive and suppress apoptosis. Limiting the drug’s bioavailability can be another way to overcome chemotherapy [46].

Recent work put the spot on cancer colon stem cell role in the development of cancer. These highly replicating cells can overexpress the ATP cassette transporters. The experimental work showed that the over-expressed MYC oncogene supports the tumor resistance by enriching ATP transporters [47]. The ATP cassette sub-family G isoform 2 is expected to overcome xenobiotic effects with abundance in the GIT and near blood vessels. Besides their role in cancer stem cells, they represent a defense mechanism against chemotherapy. Tyrosine kinase inhibitors, phosphodiesterase-5 inhibitors, and the fumitremorgin-type indolyl diketopiperazine have been used to support chemotherapy to overcome resistance by inhibiting the ATP transporters [48]. The resistance was marked in cases associated with the marker ATP cassette type G group member 2 [49]. To overcome the resistance obstacles, the use of an ATP cassette inhibitor has been introduced. Another method was to apply the new TOP-1 inhibitor FL118, which possesses the capacity to overcome efflux resistance. This new analog is weakly transported by ATP carriers [50]. KO143 is a potent antagonist of ATP cassette sub-family G member 2. It is stable and not easily metabolized by the cytochrome enzymes in the liver [51].

ATP-binding transporters have been involved in the resistance to TOP-1 inhibitors in cancer breast. Experimental work on cancer colon cell lines concluded that colon tumor cells expressed a similar mechanism. Furthermore, it is possible to add compounds inhibiting efflux mechanisms such as SCO-201 to the chemotherapy regimen to sensitize the tumor to respond to the chemotherapy indicating better survival rates [52]. It is important to notice that overexpression of the ATP transporters is a marker of resistance to cancer colon. It is recorded that candidates’ higher expression is most likely to resist camptothecin derivatives.

1.4.1 ATP efflux mechanism

The master mechanism of resistance of colorectal tumors to chemotherapy is drug efflux mechanism. The ATP-binding cassette sub-family G member 2 (ABCG2) is responsible for decreasing anti-TOP-1 bioavailability inside the tumor microenvironment leading to failure of the therapy [53]. The promoter regulating the expression of these shuttles is epigenetically regulated by methylation. Aggressive tumors express high ATP cassettes to get rid of the xenobiotic load. Hypomethylation of the promotor-regulating ATP transporter was recorded in different tumor cell lines [54]. The ABCG2 is formed from a sequence of 655 amino acids weighing 72-kilo Dalton. The structure is homodimer with two nucleotide-binding sites to export the drugs [55, 56]. The transporters play important role in the maintenance of the tumor microenvironment. They keep the balance of osmotic pressure. They have another role in antigen processing and modulation of cell division. Cell trafficking and cholesterol metabolism are affected by the ATP transporters [57].

Recently, N6-methyladenosine (m6A) RNA modification has been a new modality to treat ATP transporter through epigenetic mechanism at the RNA level [58].

Another important mechanism to defeat chemotherapy is the mutation of DNA repair response. In cancer cells, positive mutations make the tumor resistant to TOP-1 effects. The tumor genes such as BRCA1, BRCA2, PLAB2, and BARD1 have been mutated. Studies recorded alterations in 86 genes related to DNA repair. Such overactivity makes the single DNA break to the TOP-1 less effective [59].

Cancer colon cells expressing both SENP-1 (sentrin specific protease) and HIF-1α (Hypoxia inducing factor alpha) are resistant to chemotherapy. Both of them overexpress proteins SUMO pathway and make the tumor resistant to hypoxia [60]. Recently, SENP1 is a target of new compounds to develop a new regimen to overcome resistance [61].

Cancer cells can develop an alternative way to resist chemotherapy, specially irinotecan by augmenting the detoxification pathway. The active metabolite SN-38 is inactivated by glucuronidation making the drug short of killing the cancer cells. The nuclear receptors pregnane X receptors and steroid/xenobiotic receptors enhance the metabolism process of SN-38. It is important to notice that they are expressed in excess in the liver and the GIT. They also induce a battery of genes that express xenobiotic transporters that make irinotecan ineffective within a short period [62].

To repel TOP-1 inhibitors, the tumor cells induce a high copy number of the TOP-1 gene to make the drug subtherapeutic to kill the neoplastic cells. Furthermore, the TOP-1 is subjected to chromosomal alterations to be transcribed from different loci. These overexpressed loci are considered biomarkers of the drug response later on [13, 63].

The cancer colon cells are smart enough to create mutated TOP-1 that is not responding to the classic inhibitors. Experimental studies on resistant cell lines demonstrated several mutations that resist camptothecin derivatives or the new generation TOP-1 drugs [64].

The antioxidant balance in the cancer metabolism plays important role in protecting the tumor cells from xenobiotic toxicity and oxidative stress. One of the common mechanisms to support the redox is to express a high amount of glutathione reductase allowing the tumor to grow in unnatural situations and protect the mass from chemotherapy [65].

Another model for TOP-1 inhibitor resistance is colorectal tumors expressing active epidermal growth factor (EGFR). It was recorded that the active metabolite SN-38 is subjected to resistance because of the over-expression of EGFR. This factor triggers a trophic response in cancer growth via a cascade of a signaling pathway [66, 67].

1.5 Epigenetic therapy and cancer colon

New work has applied epigenetic modifier agents in combination with classic chemotherapy with a promising response. The epigenetic changes improved the drug response and reduce the needed dose to improve the clinical outcome. These transformers include DNA methyl transferase, decitabine, azacytidine, and zebularine. The study also used drugs that were considered histone deacetylase inhibitors including the well-known mood stabilizer valproic acid with promising results [68]. During the process of cancer evolution, cancer cells have different strategies to survive. It was recorded that p53 is mutated and histone deacetylases (HDACs) are over-expressed. It was considered that this epigenetic mechanism corresponds to drug resistance. Experimental inhibition of HDACs by small hairpin RNAs resulted in a better response of chemotherapy against the resistant SW480 cell lines. Indeed, epigenetic modification is part of the process of carcinogenesis and drug resistance response [69].

The DNA topology is inherited by an epigenetic mechanism. However, the process is unclear. The status of DNA twisting is a unique criterion of each cell type. This information is considered a cellular memory that is transmitted from the parent cells to the daughters by mitosis. The status of DNA 3D structure controls transcription and gene expression [70]. It has been recorded that parent cells with excess supercoiling deliver daughter cells with the impaired cell cycle. DNA supercoiling is important in the process of chromatin condensation. Previously, it was thought that DNA remains quiet during the process of mitosis. However, recent evidence that a battery of genes is still active in the mitosis process to be involved in the development of the next generation of cells [71]. The activity of the TOP-1 allows the chromatin to adapt to the DNA-positive supercoils. On the other hand, an excess of negative supercoils ends in DNA/RNA hybrid structure with resulting in DNA damage [72]. Although, DNA/RNA hybrid is considered a mechanism of regulating gene expression called an R loop [73].

1.6 Topoisomerase-1 biomarkers of response

The first line of treatment for wild-type RAS metastatic colon cancer was the anti-EGFR immunotherapy cetuximab or panitumumab [74]. Experimentation searched for the colorectal cancer biomarkers in the blood. Recently, kits to extract the tumor cDNA are available clinically to evaluate associated parameters predicting the response of the tumor to classic chemotherapy. Moreover, liquid samples assaying RAS and BRAF oncogenes have a role in chemotherapy of colorectal cancer. The assays showed that tumors with wild-type RAS and BRAF were more responsive to camptothecin chemotherapy in combination with immunotherapy as a third line of treatment for colorectal metastasis [75]. The selection of the biomarkers as a strategy of personalized medicine in colon tumors reduces the side effects for precisely predicted response to the combined TOP-1 chemotherapy-related regimen. These markers include DYPD, UGT1A1, HPP1, HER2, HER3, PIK3CA, and PTEN [76]. It is important to say that EGFR is modulating a battery-controlling factor or biomarker that can predict the tumor response. It influences RAS/BRAF/MEK/MAPK and PI3K/PTEN/AKT cascades [77].

1.7 Topoisomerase and other diseases

TOP-1 is an essential enzyme in the machinery of DNA transcription by modifying the topology of the DNA helix, and it is an important factor in stabilizing the genome. Furthermore, this enzyme has another role in the process of transcription in a way not directly related to unwinding DNA. Researchers proposed TOP-1 targets as a potential treatment for autism [78]. Mitochondria depend on the imported TOP-1 to relax the DNA during the process of transcription. Mitochondrial DNA damage is involved in many, such as neurodegenerative disorders and cancer. TOP-1 with its double role is a valuable object of cancer chemotherapy [79].

Another important scope of drugs inhibiting TOP-1 is modulating the immune response on exposure to microorganisms. Experimental studies showed that loading mice with endotoxins expressed a less harmful immune reaction than candidates tested with TOP-1 inhibitors. Low-dose TOP-1 chemotherapy such as camptothecin improved the antiviral unfavorable reaction. Interestingly, the low-dose poison with mild DNA break improved the situation in fighting the viral load [80].

Recently during the COVID-19 crisis, topotecan (TOP-1) has been applied to reduce the aberrant immune response to corona infection. Experimental work on hamsters and mice showed that a topotecan dosing improved the immune response. The TOP-1 inhibitors may exert antiviral capacity [81].

1.7.1 Mutated TOP-1

Mutated TOP-1 was a part of different chronic disorders. Malfunctioning TOP-1 is associated with unstable DNA. The causes of such complex pathology are still unknown. The hybrid TOP-1/DNA abnormal complex works as a TOP-1 inhibitor ending in excess DNA break. These phenomena were recorded in massive spinocerebellar disorders. It was reported that ATM (ataxia telangiectasia, mutated), a serine/threonine protein kinase, plays an essential role in cell cycle activation, chromatin remodeling, DNA repair, or stimulation of apoptosis. Experimental knockout of ATM showed accumulated pathogenic TOP-1/DNA complex with excess DNA break creating neurodegenerative pathology in animal models. Furthermore, healthy ATM can mediate TOP-1 complex ubiquitination [82]. These results established the idea that healthy TOP-1 is essential for development of central nervous system.

1.8 Innovations and novel strategies from a clinical perspective

Recently, the advances in multi-omic studies provided a signature of metastatic colorectal carcinoma cases aiming to predict the response of the chemotherapy. Metastatic candidates expressed unique proteomic profiles. There is a distinct profile for colorectal tumors, associated with liver metastasis. As regarding the genomic profile, there was no significant difference between early or late-stage tumors. These findings build up personalized strategies to treat or predict poor prognosis. Metastatic colorectal patients showed overexpression of oxidative phosphorylation and Krebs cycle enzymes [58]. A new strategy to use drugs that inhibit the rate of metabolism has been introduced in clinical trials as an adjuvant in colorectal metastasis in the liver. This modality is considered as tumor micro immunity reprogramming [83].

On the other hand, the analysis of circulating tumor DNA in the blood showed a mutation signature related to the tumor outcome. The status of RAS/BRAF mutations correlated with the prognosis of patients. The RAS/BRAF mutation load correlated with overall survival. As fewer mutations were detected in the blood sample, higher rates of remission were detected [84]. Another clinical study showed a similar profile. Excessive mutation RAS/BRAF suggested poor response to the first line of treatment of colorectal carcinoma [85].

A new modality mXELIRI (capecitabine plus irinotecan) has been recently applied with or without immunotherapy. The patients tolerated therapy for colorectal tumors with acceptable efficacy and toxicity profile [86]. Other trials have been applied to combine irinotecan and vincristine in soft tissue tumors [87].

New combination chemotherapy has been introduced recently. The anti-HER3 antibody patritumab has been added to the new TOP-1 inhibitor DX-8951 derivative (DXd) to treat resistant colorectal cancer with potential success [88].

Advertisement

2. Conclusions

TOP-1 gene and TOP-1 target protein are important in both carcinogenesis and pharmacology of different tumors. It is concluded that TOP-1 classic inhibitors Table 1 are the main line of treatment of metastatic colorectal carcinoma. These drugs with complex mechanism of action are associated with profound side effects and subjected to different types of machinery of drug resistance. The discovery of new drugs with multi dynamics, including mild TOP-1 in conjunction with other actions, is a promising modality to bypass the toxic effects of classic TOP-1 inhibitors as shown in Table 2. Too strong anti-TOP is no longer a wise strategy to apply because of the canonical TOP function in almost every cell. Epigenetic tools can be added to improve the chemotherapy outcome.

DrugThe studyCell lineReference
HydroxycampthecinThe drug inhibit TGF-beta1 and inhibit fibrosisHuman fibroblast[89]
Homocamptothecin E-beta ring hydroxylactoneNew anti TOP1Molecular docing[90]
1,3-disubstituted-4-hydroxy-6-methylpyridin-2(1H)-oneNew anti TOP1
Managed to treat leishmania with good cytoxicity profile
Leishmania[91]
7,12-dihydrodibenzo[b,h][1,6] naphthyridine and 7H-Chromeno[3,2-c] quinoline derivativesNon campthecin TOP1 inhibitorHuman cancer cell lines (A549 and MCF-7) and in silico[92]
9-Aminocamptothecin (9-AC)Anti TOP1 for prostate cancer with promising resultsProstate cell lines[93]
Metallated porphyrinsAnt TOP1 for different cell linesK562, U937, HL-60, Jurkat, A549 and HeLa cancer cell lines[94]
Heteroliptic cupperAnti TOP for mulriple cancer cell lines with successHuman lung (A549), cervical (HeLa) and colon (HCT-15)[95]
IrintotecanAlter metabolism of gutEnterocytes[96]

Table 1.

In vitro studies with TOP1 inhibitors.

The drugThe studyThe hostReference
Exatecan derivative (DX-8951 derivative, DXd)The drug was combined with the immunotherapy DS-8201a is a human epidermal growth factor receptor 2 (HER2)HER2-positive NCI-N87 cells and HER2-negative MDA-MB-468-Luc cells incubated in mice[97]
Topotecan (TPT)Potential role in COVID patients with success in reducing the immune stormTransgenic mice[81]
Marine alkaloid lamellarin D drivativesMore effective in treating cancer colonMurine colon cancer[98]
AZD2014 or INK128 in combination with irinotecanInduce apoptosis in Malignant peripheral nerve sheath tumorsZebra fish[99]
7-Ethyl-10-hydroxy-camptothecin (SN38), the active metabolite of irinotecan (CPT-11)Liver breast and colon cancer model expressed better responseBoth in vivo and in vitro[100]
QuercetinA flavonoid with anti TOP anti-gastric cancerBoth in vivo/in vitro[101]

Table 2.

In vivo studies with TOP1 inhibitor.

References

  1. 1. Bush NG, Evans-Roberts K, Maxwell A. DNA topoisomerases. EcoSal Plus. 2015;6:2
  2. 2. Nitiss JL, Kiianitsa K, Sun Y, Nitiss KC, Maizels N. Topoisomerase assays. Current Protocol. 2021;1(10):e250
  3. 3. Husain I, Mohler JL, Seigler HF, Besterman JM. Elevation of topoisomerase I messenger RNA, protein, and catalytic activity in human tumors: Demonstration of tumor-type specificity and implications for cancer chemotherapy. Cancer Research. 1994;54(2):539-546
  4. 4. Zhao C, Yasui K, Lee CJ, Kurioka H, Hosokawa Y, Oka T, et al. Elevated expression levels of NCOA3, TOP1, and TFAP2C in breast tumors as predictors of poor prognosis. Cancer. 2003;98(1):18-23
  5. 5. Humbert N, Martien S, Augert A, Da Costa M, Mauen S, Abbadie C, et al. A genetic screen identifies topoisomerase 1 as a regulator of senescence. Cancer Research. 2009;69(10):4101-4106
  6. 6. Bu H, Baraldo G, Lepperdinger G, Jansen-Dürr P. mir-24 activity propagates stress-induced senescence by down regulating DNA topoisomerase 1. Experimental Gerontology. 2016;75:48-52
  7. 7. Doaa G, Helaly AM. Badawi a dual pathogenesis of primary and recurrent pterygium: Immunohistochemical proof. The Open Ophthalmology. 2021;15:229-235. DOI: 10.2174/1874364102115010229
  8. 8. Liu LM, Xiong DD, Lin P, Yang H, Dang YW, Chen G. DNA topoisomerase 1 and 2A function as oncogenes in liver cancer and may be direct targets of nitidine chloride. International Journal of Oncology. 2018;53(5):1897-1912
  9. 9. Kathiravan MK, Kale AN, Nilewar S. Discovery and development of topoisomerase inhibitors as anticancer agents. Mini Reviews in Medicinal Chemistry. 2016;16(15):1219-1229
  10. 10. Nukuzuma S, Nakamichi K, Kameoka M, Sugiura S, Nukuzuma C, Tasaki T, et al. Suppressive effect of topoisomerase inhibitors on JC polyomavirus propagation in human neuroblastoma cells. Microbiology and Immunology. 2016;60(4):253-260
  11. 11. Li M, Pokharel S, Wang JT, Xu X, Liu Y. RECQ5-dependent SUMOylation of DNA topoisomerase I prevents transcription-associated genome instability. Nature Communications. 2015;6:6720
  12. 12. Han ZJ, Feng YH, Gu BH, Li YM, Chen H. The post-translational modification, SUMOylation, and cancer (Review). International Journal of Oncology. 2018;52(4):1081-1094
  13. 13. Braun MS, Richman SD, Quirke P, Daly C, Adlard JW, Elliott F, et al. Predictive biomarkers of chemotherapy efficacy in colorectal cancer: Results from the UK MRC FOCUS trial. Journal of Clinical Oncology. 2008a;26(16):2690-2698
  14. 14. Marx C, Sonnemann J, Beyer M, Maddocks ODK, Lilla S, Hauzenberger I, et al. Mechanistic insights into p53-regulated cytotoxicity of combined entinostat and irinotecan against colorectal cancer cells. Molecular in Oncology. 2021;15(12):3404-3429
  15. 15. Marx C, Sonnemann J, Maddocks ODK, Marx-Blümel L, Beyer M, Hoelzer D, et al. Global metabolic alterations in colorectal cancer cells during irinotecan-induced DNA replication stress. Cancer Metabolism. 2022;10(1):10
  16. 16. Wall ME, Wani MC. Camptothecin and taxol: Discovery to clinic--thirteenth Bruce F. Cancer Research. 1995;55(4):753-760
  17. 17. Martín-Encinas E, Selas A, Palacios F, Alonso C. The design and discovery of topoisomerase I inhibitors as anticancer therapies. Expert Opinion in Drug Discovery. 2022;17(6):581-601
  18. 18. Talukdar A, Kundu B, Sarkar D, Goon S, Mondal MA. Topoisomerase I inhibitors: Challenges, progress and the road ahead. European Journal of Medicinal Chemistry. 2022;236:114304
  19. 19. Rasheed ZA, Rubin EH. Mechanisms of resistance to topoisomerase I-targeting drugs. Oncogene. 2003;22(47):7296-7304
  20. 20. O'Connor MJ. Targeting the DNA damage response in Cancer. Molecular Cell. 2015;60(4):547-560
  21. 21. Pommier Y. Topoisomerase I inhibitors: Camptothecins and beyond. Nature Reviews. Cancer. 2006;6(10):789-802
  22. 22. Xu Y, Her C. Inhibition of topoisomerase (DNA) I (TOP1): DNA damage repair and anticancer therapy. Biomolecules. 2015;5(3):1652-1670
  23. 23. Cinelli MA. Topoisomerase 1B poisons: Over a half-century of drug leads, clinical candidates, and serendipitous discoveries. Medicinal Research Reviews. 2019;39(4):1294-1337
  24. 24. Guichard S, Terret C, Hennebelle I, Lochon I, Chevreau P, Frétigny E, et al. CPT-11 converting carboxylesterase and topoisomerase activities in tumour and normal colon and liver tissues. British Journal of Cancer. 1999;80(3-4):364-370
  25. 25. Mathijssen RH, van Alphen RJ, Verweij J, Loos WJ, Nooter K, Stoter G, et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clinical Cancer Research. 2001;7(8):2182-2194
  26. 26. Slatter JG, Schaaf LJ, Sams JP, Feenstra KL, Johnson MG, Bombardt PA, et al. Pharmacokinetics, metabolism, and excretion of irinotecan (CPT-11) following I.V. infusion of [(14)C]CPT-11 in cancer patients. Drug Metabolism and Disposition. 2000;28(4):423-433
  27. 27. Bailly C. Irinotecan: 25 years of cancer treatment. Pharmacological Research. 2019;148:104398
  28. 28. Li F, Jiang T, Li Q , Ling X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: Did we miss something in CPT analogue molecular targets for treating human disease such as cancer? American Journal of Cancer Research. 2017;7(12):2350-2394
  29. 29. Covey JM, Jaxel C, Kohn KW, Pommier Y. Protein-linked DNA strand breaks induced in mammalian cells by camptothecin, an inhibitor of topoisomerase I. Cancer Research. 1989;49(18):5016-5022
  30. 30. Thomas A, Pommier Y. Targeting topoisomerase I in the era of precision medicine. Clinical Cancer Research. 2019;25(22):6581-6589
  31. 31. Hur J, Ghosh M, Kim TH, Park N, Pandey K, Cho YB, et al. Synergism of AZD6738, an ATR inhibitor, in combination with belotecan, a Camptothecin analogue, in chemotherapy-resistant ovarian Cancer. International Journal of Molecular Sciences. 2021;22(3):1223
  32. 32. Jun E, Park Y, Lee W, Kwon J, Lee S, Kim MB, et al. The identification of candidate effective combination regimens for pancreatic cancer using the histoculture drug response assay. Scientific Reports. 2020;10(1):12004
  33. 33. Kang JH, Lee KH, Kim DW, Kim SW, Kim HR, Kim JH, et al. A randomised phase 2b study comparing the efficacy and safety of belotecan vs. topotecan as monotherapy for sensitive-relapsed small-cell lung cancer. British Journal of Cancer. 2021;124(4):713-720
  34. 34. Cesare DE. High efficacy of intravenous Gimatecan on human tumor xenografts. Anticancer Research. 2018;38(10):5783-5790
  35. 35. Kasi A, McGinnis T, Naik G, Handa S, Williams G, Paluri R. Efficacy and tolerability of the combination of nano-liposomal irinotecan and 5-fluorouracil/leucovorin in advanced pancreatic adenocarcinoma: Post-approval clinic experience. Journal of Gastrointestinal Oncology. 2021;12(2):464-473
  36. 36. Yasuoka H, Naganuma A, Kurihara E, Kobatake T, Ijima M, Tamura Y, et al. Efficacy and safety of the combination of Nano-liposomal irinotecan and 5-fluorouracil/L-leucovorin in unresectable advanced pancreatic cancer: A real-world study. Oncology. 2022;100(8):449-459
  37. 37. Liu X, Jiang J, Chan R, Ji Y, Lu J, Liao YP, et al. Improved efficacy and reduced toxicity using a custom-designed irinotecan-delivering silicasome for orthotopic colon cancer. ACS Nano. 2019;13(1):38-53
  38. 38. Xing J, Zhang X, Wang Z, Zhang H, Chen P, Zhou G, et al. Novel lipophilic SN38 prodrug forming stable liposomes for colorectal carcinoma therapy. International Journal of Nanomedicine. 2019;14:5201-5213
  39. 39. Gokduman K. Strategies targeting DNA topoisomerase I in Cancer chemotherapy: Camptothecins, nanocarriers for camptothecins, organic non-Camptothecin compounds and metal complexes. Current Drug Targets. 2016;17(16):1928-1939
  40. 40. Cushman M. Design and synthesis of Indenoisoquinolines targeting topoisomerase I and other biological macromolecules for Cancer chemotherapy. Journal of Medicinal Chemistry. 2021;64(24):17572-17600
  41. 41. Wang KB, Elsayed MSA, Wu G, Deng N, Cushman M, Yang D. Indenoisoquinoline topoisomerase inhibitors strongly bind and stabilize the MYC promoter G-Quadruplex and downregulate MYC. Journal of American Chemical Society. 2019;141(28):11059-11070
  42. 42. Cho WJ, Park MJ, Imanishi T, Chung BH. A novel synthesis of benzo[c]phenanthridine skeleton and biological evaluation of isoquinoline derivatives. Chemical Pharmacy Bulletin (Tokyo). 1999;47(6):900-902
  43. 43. Khadka DB, Le QM, Yang SH, Van HT, Le TN, Cho SH, et al. Design, synthesis and docking study of 5-amino substituted indeno[1,2-c]isoquinolines as novel topoisomerase I inhibitors. Bioorganic & Medicinal Chemistry. 2011;19(6):1924-1929
  44. 44. Strumberg D, Pommier Y, Paull K, Jayaraman M, Nagafuji P, Cushman M. Synthesis of cytotoxic indenoisoquinoline topoisomerase I poisons. Journal of Medicinal Chemistry. 1999;42(3):446-457
  45. 45. Park EJ, Kiselev E, Conda-Sheridan M, Cushman M, Pezzuto JM. Induction of apoptosis by 3-amino-6-(3-aminopropyl)-5,6-dihydro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline via modulation of MAPKs (p38 and c-Jun N-terminal kinase) and c-Myc in HL-60 human leukemia cells. Journal of Natural Products. 2012;75(3):378-384
  46. 46. Tomicic MT, Kaina B. Topoisomerase degradation, DSB repair, p53 and IAPs in cancer cell resistance to camptothecin-like topoisomerase I inhibitors. Biochimica et Biophysica Acta. 2013;1835(1):11-27
  47. 47. Zhang HL, Wang P, Lu MZ, Zhang SD, Zheng L. c-Myc maintains the self-renewal and chemoresistance properties of colon cancer stem cells. Oncology Letters. 2019;17(5):4487-4493
  48. 48. Stacy AE, Jansson PJ, Richardson DR. Molecular pharmacology of ABCG2 and its role in chemoresistance. Molecular Pharmacology. 2013;84(5):655-669. DOI: 10.1124/mol.113.088609
  49. 49. Tuy HD, Shiomi H, Mukaisho KI, Naka S, Shimizu T, Sonoda H, et al. ABCG2 expression in colorectal adenocarcinomas may predict resistance to irinotecan. Biochimica et Biophysica Acta. 2016;12(4):2752-2760
  50. 50. Westover D, Ling X, Lam H, Welch J, Jin C, Gongora C, et al. FL118, a novel camptothecin derivative, is insensitive to ABCG2 expression and shows improved efficacy in comparison with irinotecan in colon and lung cancer models with ABCG2-induced resistance. Molecular Cancer. 2015;14:92
  51. 51. Liu K, Zhu J, Huang Y, Li C, Lu J, Sachar M, et al. Metabolism of KO143, an ABCG2 inhibitor. Drug Metabolism and Pharmacokinetics. 2017;32(4):193-200
  52. 52. Ambjørner SEB, Wiese M, Köhler SC, Svindt J, Lund XL, Gajhede M, et al. The pyrazolo[3,4-d]pyrimidine derivative, SCO-201, reverses multidrug resistance mediated by ABCG2/BCRP. Cell. 2020;9(3):613
  53. 53. Moon HH, Kim SH, Ku JL. Correlation between the promoter methylation status of ATP-binding cassette sub-family G member 2 and drug sensitivity in colorectal cancer cell lines. Oncology Reports. 2016;35(1):298-306
  54. 54. Spitzwieser M, Pirker C, Koblmüller B, Pfeiler G, Hacker S, Berger W, et al. Promoter methylation patterns of ABCB1, ABCC1 and ABCG2 in human cancer cell lines, multidrug-resistant cell models and tumor, tumor-adjacent and tumor-distant tissues from breast cancer patients. Oncotarget. 2016;7(45):73347-73369
  55. 55. Litman T, Brangi M, Hudson E, Fetsch P, Abati A, Ross DD, et al. The multidrug-resistant phenotype associated with overexpression of the new ABC half-transporter, MXR (ABCG2). Journal of Cell Science. 2000;113(Pt 11):2011-2021
  56. 56. Xu J, Liu Y, Yang Y, Bates S, Zhang JT. Characterization of oligomeric human half-ABC transporter ATP-binding cassette G2. The Journal of Biological Chemistry. 2004;279(19):19781-19789
  57. 57. Liu X. ABC family transporters. Advances in Experimental Medicine and Biology. 2019;1141:13-100
  58. 58. Li C, Sun YD, Yu GY, Cui JR, Lou Z, Zhang H, et al. Integrated omics of metastatic colorectal Cancer. Cancer Cell. 2020;38(5):734-747
  59. 59. Cuella-Martin R, Hayward SB, Fan X, Chen X, Huang JW, Taglialatela A, et al. Functional interrogation of DNA damage response variants with base editing screens. Cell. 2021;184(4):1081-1097
  60. 60. Chen MC, Nhan DC, Hsu CH, Wang TF, Li CC, Ho TJ, et al. SENP1 participates in irinotecan resistance in human colon cancer cells. Journal of Cellular Biochemistry. 2021;122(10):1277-1294
  61. 61. Wei J, Wang H, Zheng Q , Zhang J, Chen Z, Wang J, et al. Recent research and development of inhibitors targeting sentrin-specific protease 1 for the treatment of cancers. European Journal of Medicinal Chemistry. 2022;241:114650
  62. 62. Ozawa S, Miura T, Terashima J, Habano W. Cellular irinotecan resistance in colorectal cancer and overcoming irinotecan refractoriness through various combination trials including DNA methyltransferase inhibitors: A review. Cancer Drug Resistant. 2021;4(4):946-964
  63. 63. Rømer MU, Jensen NF, Nielsen SL, Müller S, Nielsen KV, Nielsen HJ, et al. TOP1 gene copy numbers in colorectal cancer samples and cell lines and their association to in vitro drug sensitivity. Scandinavian Journal of Gastroenterology. 2012;47(1):68-79
  64. 64. Jensen NF, Agama K, Roy A, Smith DH, Pfister TD, Rømer MU, et al. Characterization of DNA topoisomerase I in three SN-38 resistant human colon cancer cell lines reveals a new pair of resistance-associated mutations. Journal of Experimental & Clinical Cancer Research. 2016;35:56
  65. 65. Narayanankutty A, Job JT, Narayanankutty V. Glutathione, an antioxidant tripeptide: Dual roles in carcinogenesis and chemoprevention. Current Protein & Peptide Science. 2019;20(9):907-917
  66. 66. Asano T. Drug resistance in cancer therapy and the role of epigenetics. Journal of Nippon Medical School. 2020;87(5):244-251
  67. 67. Petitprez A, Larsen AK. Irinotecan resistance is accompanied by upregulation of EGFR and Src signaling in human cancer models. Current Pharmaceutical Design. 2013;19(5):958-964
  68. 68. Hosokawa M, Tanaka S, Ueda K, Iwakawa S. Different schedule-dependent effects of epigenetic modifiers on cytotoxicity by anticancer drugs in colorectal Cancer cells. Biological & Pharmaceutical Bulletin. 2017;40(12):2199-2204
  69. 69. Alzoubi S, Brody L, Rahman S, Mahul-Mellier AL, Mercado N, Ito K, et al. Synergy between histone deacetylase inhibitors and DNA-damaging agents is mediated by histone deacetylase 2 in colorectal cancer. Oncotarget. 2016;7(28):44505-44521
  70. 70. Jha RK, Levens D, Kouzine F. Mechanical determinants of chromatin topology and gene expression. Nucleus. 2022;13(1):94-115
  71. 71. Palozola KC, Donahue G, Liu H, Grant GR, Becker JS, Cote A, et al. Mitotic transcription and waves of gene reactivation during mitotic exit. Science. 2017;358(6359):119-122
  72. 72. Edwards DS, Maganti R, Tanksley JP, Luo J, Park JJH, Balkanska-Sinclair E, et al. BRD4 prevents R-loop formation and transcription-replication conflicts by ensuring efficient transcription elongation. Cell Reports. 2020;32(12):108166
  73. 73. Yuan W, Al-Hadid Q , Wang Z, Shen L, Cho H, Wu X, et al. TDRD3 promotes DHX9 chromatin recruitment and R-loop resolution. Nucleic Acids Research. 2021;49(15):8573-8591
  74. 74. Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Annals of Oncology. 2016;27(8):1386-1422
  75. 75. Cremolini C, Rossini D, Dell'Aquila E, Lonardi S, Conca E, Del Re M, et al. Rechallenge for patients with RAS and BRAF wild-type metastatic colorectal cancer with acquired resistance to first-line cetuximab and irinotecan: A phase 2 single-arm clinical trial. JAMA Oncology. 2019;5(3):343-350
  76. 76. Taieb J, Jung A, Sartore-Bianchi A, Peeters M, Seligmann J, Zaanan A, et al. The evolving biomarker landscape for treatment selection in metastatic colorectal Cancer. Drugs. 2019;79(13):1375-1394
  77. 77. Peeters M, Price TJ, Cervantes A, Sobrero AF, Ducreux M, Hotko Y, et al. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. Journal of Clinical Oncology. 2010;28(31):4706-4713
  78. 78. Li M, Liu Y. Topoisomerase I in human disease pathogenesis and treatments. Genomics Proteomics Bioinformatics. 2016;14(3):166-171
  79. 79. Das BB, Ghosh A, Bhattacharjee S, Bhattacharyya A. Trapped topoisomerase-DNA covalent complexes in the mitochondria and their role in human diseases. Mitochondrion. 2021;60:234-244
  80. 80. Pépin G, Nejad C, Ferrand J, Thomas BJ, Stunden HJ, Sanij E, et al. Topoisomerase 1 inhibition promotes cyclic GMP-AMP synthase-dependent antiviral responses. MBio. 2017;8(5):e01611-e01617
  81. 81. Ho JSY, Mok BW, Campisi L, Jordan T, Yildiz S, Parameswaran S, et al. TOP1 inhibition therapy protects against SARS-CoV-2-induced lethal inflammation. Cell. 2021;184(10):2618-2632
  82. 82. Katyal S, Lee Y, Nitiss KC, Downing SM, Li Y, Shimada M, et al. Aberrant topoisomerase-1 DNA lesions are pathogenic in neurodegenerative genome instability syndromes. Nature Neuroscience. 2014;17(6):813-821
  83. 83. Wu Y, Yang S, Ma J, Chen Z, Song G, Rao D, et al. Spatiotemporal immune landscape of colorectal Cancer liver metastasis at single-cell level. Cancer Discovery. 2022;12(1):134-153
  84. 84. Wang F, Huang YS, Wu HX, Wang ZX, Jin Y, Yao YC, et al. Genomic temporal heterogeneity of circulating tumour DNA in unresectable metastatic colorectal cancer under first-line treatment. Gut. 2022;71(7):1340-1349
  85. 85. Yao J, Zang W, Ge Y, Weygant N, Yu P, Li L, et al. RAS/BRAF circulating tumor DNA mutations as a predictor of response to first-line chemotherapy in metastatic colorectal Cancer patients. Canadian Journal of Gastroenterology & Hepatology. 2018;2018:4248971
  86. 86. Xu RH, Muro K, Morita S, et al. Modified XELIRI (capecitabine plus irinotecan) versus FOLFIRI (leucovorin, fluorouracil, and irinotecan), both either with or without bevacizumab, as second-line therapy for metastatic colorectal cancer (AXEPT): A multicentre, open-label, randomised, non-inferiority, phase 3 trial. The Lancet Oncology. 2018;19(5):660-671. DOI:10.1016/S1470-2045(18)30140-2
  87. 87. Defachelles AS, Bogart E, Casanova M, Merks JHM, Bisogno G, Calareso G, et al. Randomized phase II trial of vincristine-irinotecan with or without temozolomide, in children and adults with relapsed or refractory rhabdomyosarcoma: A European paediatric soft tissue sarcoma study group and innovative therapies for children with cancer trial. Journal of Clinical Oncology. 2021;39(27):2979-2990
  88. 88. Koganemaru S, Kuboki Y, Koga Y, Kojima T, Yamauchi M, Maeda N, et al. U3-1402, a novel HER3-targeting antibody-drug conjugate, for the treatment of colorectal Cancer. Molecular Cancer Therapeutics. 2019;18(11):2043-2050
  89. 89. Yao Z, Zheng W, Zhang X, Xiong H, Qian Y, Fan C. Hydroxycamptothecin prevents fibrotic pathways in fibroblasts In vitro. IUBMB Life. 2019;71(5):653-662
  90. 90. Laco GS, Du W, Kohlhagen G, Sayer JM, Jerina DM, Burke TG, et al. Analysis of human topoisomerase I inhibition and interaction with the cleavage site +1 deoxyguanosine, via in vitro experiments and molecular modeling studies. Bioorganic & Medicinal Chemistry. 2004;12(19):5225-5235
  91. 91. Borkar MR, Martis EAF, Nandan S, Patil RH, Shelar A, Iyer KR, et al. Identification of potential antileishmanial 1,3-disubstituted-4-hydroxy-6-methylpyridin-2(1H)-ones, in vitro metabolic stability, cytotoxicity and molecular modeling studies. Chemico-Biological Interactions. 2022 Jan;5(351):109758
  92. 92. Kardile RA, Sarkate AP, Borude AS, Mane RS, Lokwani DK, Tiwari SV, et al. Design and synthesis of novel conformationally constrained 7,12-dihydrodibenzo[b,h][1,6] naphthyridine and 7H-Chromeno[3,2-c] quinoline derivatives as topoisomerase I inhibitors: In vitro screening, molecular docking and ADME predictions. Bioorganic Chemistry. 2021;115:105174
  93. 93. de Souza PL, Cooper MR, Imondi AR, Myers CE. 9-Aminocamptothecin: A topoisomerase I inhibitor with preclinical activity in prostate cancer. Clinical Cancer Research. 1997;3(2):287-294
  94. 94. Halder N, Dzhemileva LU, Ramazanov IR, D'yakonov VA, Dzhemilev UM, Rath H. Comparative in vitro studies of the topoisomerase I inhibition and anticancer activities of metallated N-confused porphyrins and metallated porphyrins. ChemMedChem. 2020;15(7):632-642
  95. 95. Haleel A, Mahendiran D, Veena V, Sakthivel N, Rahiman AK. Antioxidant, DNA interaction, VEGFR2 kinase, topoisomerase I and in vitro cytotoxic activities of heteroleptic copper(II) complexes of tetrazolo[1,5-a]pyrimidines and diimines. Materials Science & Engineering. C, Materials for Biological Applications. 2016;68:366-382
  96. 96. Parvez MM, Basit A, Jariwala PB, Gáborik Z, Kis E, Heyward S, et al. Quantitative investigation of irinotecan metabolism, transport, and gut microbiome activation. Drug Metabolism and Disposition. 2021;49(8):683-693
  97. 97. Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Science. 2016;107(7):1039-1046
  98. 98. Fukuda T, Nanjo Y, Fujimoto M, Yoshida K, Natsui Y, Ishibashi F, et al. Lamellarin-inspired potent topoisomerase I inhibitors with the unprecedented benzo[g][1]benzopyrano[4,3-b]indol-6(13H)-one scaffold. Bioorganic & Medicinal Chemistry. 2019;27(2):265-277
  99. 99. Ki DH, Oppel F, Durbin AD, Look AT. Mechanisms underlying synergy between DNA topoisomerase I-targeted drugs and mTOR kinase inhibitors in NF1-associated malignant peripheral nerve sheath tumors. Oncogene. 2019;38(39):6585-6598
  100. 100. Liu H, Lu H, Liao L, Zhang X, Gong T, Zhang Z. Lipid nanoparticles loaded with 7-ethyl-10-hydroxycamptothecin-phospholipid complex: in vitro and in vivo studies. Drug Delivery. 2015;22(6):701-709
  101. 101. Lei CS, Hou YC, Pai MH, Lin MT, Yeh SL. Effects of quercetin combined with anticancer drugs on metastasis-associated factors of gastric cancer cells: in vitro and in vivo studies. The Journal of Nutritional Biochemistry. 2018;51:105-113

Written By

Ahmed Mohamed Nabil Helaly and Doaa Ghorab

Submitted: 19 October 2022 Reviewed: 10 November 2022 Published: 22 December 2022