Next Article in Journal
Recent Achievements in Electrochemical and Optical Nucleic Acids Based Detection of Metal Ions
Previous Article in Journal
Medicinal Values and Potential Risks Evaluation of Ginkgo biloba Leaf Extract (GBE) Drinks Made from the Leaves in Autumn as Dietary Supplements
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Breaking the Bottleneck in Anticancer Drug Development: Efficient Utilization of Synthetic Biology

1
Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
2
Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(21), 7480; https://doi.org/10.3390/molecules27217480
Submission received: 6 September 2022 / Revised: 20 October 2022 / Accepted: 24 October 2022 / Published: 2 November 2022

Abstract

:
Natural products have multifarious bioactivities against bacteria, fungi, viruses, cancers and other diseases due to their diverse structures. Nearly 65% of anticancer drugs are natural products or their derivatives. Thus, natural products play significant roles in clinical cancer therapy. With the development of biosynthetic technologies, an increasing number of natural products have been discovered and developed as candidates for clinical cancer therapy. Here, we aim to summarize the anticancer natural products approved from 1950 to 2021 and discuss their molecular mechanisms. We also describe the available synthetic biology tools and highlight their applications in the development of natural products.

Graphical Abstract

1. Introduction

Cancer is one of the deadliest diseases, in which cell grow uncontrollably and then are shed into the blood, which transports the cells to other parts of the body. Cancer has been highlighted as one of the most serious health challenges worldwide. According to global cancer statistics, in 2020, 19.3 million new cancer cases and 10.0 million cancer deaths were reported [1]. In particular, in 2020, the cumulative risk of cancer incidence and mortality increased to 20.58% and 10.73%, which are 2.08 and 1.90 times higher than these in 2002, respectively [2]. Thus, an increasing number of researchers are focusing on cancer research and therapy [3,4]. Conventional cancer therapies include surgery, radiation, and chemotherapy. Clinical cancer drugs play essential roles in cancer therapy. However, chemotherapy is often accompanied by the emergence of resistance, cancer recurrence and the development of serious side effects, such as peripheral neuropathy, nephropathy and liver injury [5,6]. Therefore, there remains an urgent demand for clinical drugs with excellent curative effects against cancer.
Natural products (NPs) originate from animals, plant extracts, insects, marine organisms, microorganisms and other organisms. They are produced by a series of mechanisms in hosts, mainly including polyketide synthase (PKS), nonribosomal peptide synthetase (NRPS) and ribosomally synthesized and posttranslationally modified peptide (RiPP) mechanisms. Due to the wide variety of biosynthetic mechanisms, NPs exhibit diverse structures, biological activities and applications, especially in the treatment of human diseases and in veterinary and agricultural applications [7]. As primary and secondary metabolites or intermediates, NPs do not involve dangerous chemical synthesis processes and byproducts.
NPs, such as the anticancer drug paclitaxel [8], immunosuppressive drug rapamycin [9] and antiparasitic drug artemisinin [10], have been extensively applied for the prevention and treatment of various human diseases. As of 2019, 64.4% of the 1881 drugs approved by the United States Food and Drug Administration (U.S. FDA) are NPs or their derivatives and analogues, such as plant-derived artemisinin and microorganism-derived fidaxomicin [11]. At present, nearly 70% of antibacterial drugs and 65% of antitumor drugs come from NPs and their derivatives [12], such as daptomycin [13] and actinomycin. Due to their structural diversity, NPs have a broad array of anticancer mechanisms. They can inhibit cancer development by arresting cell proliferation and promoting cell apoptosis, autophagy and immunotherapy via a series of signaling pathways [14,15].
To discover novel and valuable NPs, an increasing number of strategies and tools have been developed and applied. Synthetic biology is the most popular and powerful strategy for discovering novel and valuable NPs. As an emerging field of biological research in the early 21st century, synthetic biology aims to artificially design and construct new biological systems with specific physiological functions to establish biological manufacturing pathways for products, such as drugs, functional materials or energy substitutes. To design and construct biological systems, numerous tools and methods have been developed, such as the antibiotics and secondary metabolite analysis shell (antiSMASH) web server [16], clustered regularly interspaced short palindromic repeats-CRISPR associated protein (CRISPR-Cas) [17,18], DNA assembly [19], direct cloning [20], transcriptional regulation [21], protein engineering [22], multiple omics [23] and host engineering [24].
In this review, we will summarize the approved anticancer natural drugs covering from 1950 to 2021 and discuss the great potential of the clinical applications of NPs in cancer therapy and summarize their anticancer mechanisms. We will also outline the synthetic biology strategies and tools used for discovering valuable NPs and highlight their applications in NP development.

2. FDA-Approved Anticancer Natural Products

As a typical and general medical therapy for cancer, chemotherapeutic drugs can also damage normal cells and then cause a series of complications. Moreover, drug resistance emerges with the wide use of chemotherapeutic drugs, especially in advanced-stage cancers. All these factors can result in the failure of cancer therapy. Thus, there is a need for drugs with strong activities against cancer cells and low toxicity against normal cells. Different NPs, on the one hand, exhibit effective activities against various types of cancer; on the other hand, they play prominent roles in drug resistance via various mechanisms [25,26,27]. Moreover, they can also improve the effects of immune therapy [15]. In brief, natural products are becoming significant for medicine research.
In this section, we summarize all FDA-approved anticancer NPs, covering 72 years from 1950 to 2021. In total, 87 NPs are summarized and discussed according to their names and sources (Table 1). Forty-five percent of anticancer drugs are derived from large biological macromolecules, NPs or their derivatives (Figure 1A). Specifically, the number of NPs applied in cancer therapy has grown rapidly from 1950 to 2020 (Figure 1B, Table 1). In the 1950s, 7 NPs were approved for application in clinical cancer therapy. Meanwhile, in the 2010s, 21 NPs were approved for cancer therapy, three times the number approved in the 1950s. The anticancer cancer NPs approved in the last two decades account for 41% of all approved NPs (Figure 1C). This suggests that with the development of technology, an increasing number of NPs are being found and applied in clinical cancer therapy. More interestingly, the number of steroids decreased gradually, and only one hormone was approved in the last decade, for therapy of castration-resistant prostate cancer, which is four times lower than the number approved in the 1950s (Figure 1D, Table 1). On the other hand, marine NPs and antibody drug conjugates (ADCs) have emerged and increased rapidly in the last two decades, and they will be the main sources of clinical drugs. Except for these approved NPs, a large number of NPs have been reported to be toxic to cancer cells through various mechanisms, and some of them entered clinical trials [28,29,30]. These unapproved NPs still have great potential as clinical drugs.

3. Anticancer Mechanisms of Natural Products

As we know, human cancer is produced from multiple processes. The cancer cells in different processes seem to exhibit different functional capabilities. Hanahan D. envisages eight distinct hallmark capabilities, including evading growth suppress, avoiding immune destruction, enabling replicative immortality, activating invasion and metastasis, inducing or accessing vasculature and so on, to rationalize the complex phenotypes of diverse human tumor types and variants [118]. Destroying the hallmark capabilities in tumor progresses is promising for tumor suppression.
NPs suppress cancer development via diverse anticancer mechanisms due to their various chemical structures (Figure 2). For example, the intact lactone ring of camptothecins and the colchicine domain of podophyllotoxins are widely recognized as the active structures for their anticancer activities [119,120]. Although the antitumor effects of some NPs have been reviewed previously [14,121], the molecular mechanisms of NPs in cancer development have not been comprehensively summarized and discussed. Thus, we further summarize the molecular mechanisms of NPs in tumor suppression in this section.

3.1. Plant-Derived Drugs

Most natural anticancer drugs identified to date are plant-derived NPs, mainly derived from paclitaxel, camptothecin, podophyllotoxin, vinca-alkaloid and their analogues. Due to the numerous types of plant-derived drugs, their anticancer mechanisms are varied.
Microtubules play significant roles in the proliferation, migration and invasion of cancer cells. Various anticancer agents have been designed to target microtubules. There are two effective antimicrotubule therapeutics, paclitaxel and vinca-alkaloids. Paclitaxel and its analogues bind to tubulin and then promote tubulin polymerization and inhibit microtubule disassembly [122]. In contrast to paclitaxel, vinca-alkaloids and their analogues, such as vinflunine, vinblastine and vincristine, inhibit tubulin polymerization and subsequent spindle assembly [123] (Figure 3A). Microtubule dynamics change dramatically over time, which is essential for promoting microtubule growth and disassembly. Both of these classes of drugs induce the failure of microtubule dynamics. Microtubule dynamics instability is highly responsible for mitotic chromosome instability, which is common in cancer cells.
Camptothecin analogues, including belotecan HCl, topotecan HCl and irinotecan HCl, kill cancer cells by specifically targeting and binding to DNA topoisomerase I, which results in a double-stranded DNA break during the S-phase [124]. Another class of NPs, podophyllotoxin analogues, have been identified as topoisomerase II inhibitors. The most recent reports suggest that etoposide and its metabolites interact with topoisomerase II enzymes on the one hand, and they can also covalently bind to CREB-binding protein and T cell protein tyrosine phosphatase on the other hand [54]. These two proteins are functional in the differentiation and proliferation of hematopoietic stem cells. The binding of etoposide directly results in enzyme inhibition. DNA topoisomerases are essential in DNA replication, transcription and chromosome segregation. Their inhibitors block these biological processes and then disrupt cell proliferation.
In addition, some other plant NPs, such as homoharringtonine, arglabin and solamargines, suppress tumor growth via specific pathways (Figure 3B). For example, arglabin and solamargines induce apoptosis via the mitochondrial pathway, which is one of the major mechanisms of apoptosis leading to programmed cell death. In this pathway, upon stimulation by apoptosis signal(s), cytochrome c is released from mitochondria to the cytoplasm and then interacts with Apaf-1 to form apoptotic bodies. The structurally changed Apaf-1 in the apoptotic body recruits caspase 9 and causes caspase 9 to be cleaved into two segments and activated. Activated caspase 9 further activates subsequent caspase proteins and then initiates apoptosis. Arglabin and solamargines inhibit the activity of Bcl-2 and upregulate caspase-3, respectively [125,126].
Figure 3. The main molecular mechanisms and regulation networks of anticancer drugs derived from natural products. (A) Natural products inhibit cell proliferation by destroying the microtubule dynamic and DNA damage and inhibiting translation. (B) Natural products induce apoptosis via mitochondrial pathway and chromatin damage and by destroying the balance of HAT and HDAC. (C) Biosynthesis of doxorubicin, a type II polyketide [127]. ACP: acyl carrier protein. KS: ketosynthase. KR: ketoreductase. GT: glycosyltransferase. MT: methyl transferase. OA: oxygenase. ME: methyl esterase. (D) Natural products result in vascular shutdown via Rho pathway. ES: endoplasmic reticulum stress. OS: oxidative stress. HAT: histone acetylase. HDAC: histone deacetylase. IFP: incorrectly folded protein. TOP: topoisomerase.
Figure 3. The main molecular mechanisms and regulation networks of anticancer drugs derived from natural products. (A) Natural products inhibit cell proliferation by destroying the microtubule dynamic and DNA damage and inhibiting translation. (B) Natural products induce apoptosis via mitochondrial pathway and chromatin damage and by destroying the balance of HAT and HDAC. (C) Biosynthesis of doxorubicin, a type II polyketide [127]. ACP: acyl carrier protein. KS: ketosynthase. KR: ketoreductase. GT: glycosyltransferase. MT: methyl transferase. OA: oxygenase. ME: methyl esterase. (D) Natural products result in vascular shutdown via Rho pathway. ES: endoplasmic reticulum stress. OS: oxidative stress. HAT: histone acetylase. HDAC: histone deacetylase. IFP: incorrectly folded protein. TOP: topoisomerase.
Molecules 27 07480 g003

3.2. Microorganism-Derived Drugs

Streptomyces strains are the main sources of microorganism-derived drugs. Although the genetic manipulation and fermentation of Streptomyces are time-consuming, these organisms possess rich precursors, cofactors and posttranslational modifications, which are essential for the diverse bioactivities of natural products [24]. Due to the diverse structures of microorganism-derived drugs, their mechanisms vary.
Nearly half of microorganism-derived drugs are anthracyclines, including aclarubicin, daunomycin, doxorubicin and their derivatives. They are synthesized via type II polyketide pathways [127] (Figure 3C). As the most effective curative regimen for a series of hematologic cancers and solid cancers, there are two important mechanisms by which doxymycin, daunomycin, epirubicin and idarubicin induce cancer cell apoptosis: DNA damage and chromatin damage [128] (Figure 3B). On the one hand, anthracyclines are composed of tetracycline aglycones related to aminoglycosides. The amino sugar inserts into the DNA minor groove, while the tetracyclic moiety inserts into the DNA double helix [129]. This action induces topoisomerase II poisoning and subsequent DNA double-strand breaks (DSBs). On the other hand, anthracyclines release histones from the genome, which results in chromatin damage and epigenomic and transcriptional effects. However, the combination of DNA and chromatin damage contributes to serious side effects, such as cardiovascular toxicity [130] and infertility [131]. Furthermore, as an analogue of doxymycin, aclarubicin evicts histones from the genome without inducing DSBs. It shows effective anticancer activity but less toxicity than other anthracyclines [131]. These results provide strategies with which researchers can improve the activity and safety of anthracycline drugs.
Additionally, bleomycin and mitomycin C preferentially cleave actively transcribed genes, and telomeric DNA is their major target. In contrast, bleomycin produces free radicals that act on chromosomes and then induce chromosomal aberrations [132]. Romidepsin is produced from by Chromobacterium violaceum and disrupts the balance of histone deacetylase by inhibiting the activity of class I histone deacetylase enzymes in refractory or relapsed cutaneous and peripheral T cell lymphomas (Figure 3B). Transcriptional suppression is another anticancer mechanism of microorganism-derived drugs, such as actinomycin D and mithramycin. Actinomycin D is an RNA polymerase inhibitor, and mithramycin is an inhibitor of Sp-1, which is a transcription factor of ACVRL-1 [75]. Temsirolimus is a derivative of sirolimus produced by Streptomyces hygroscopicus. It is used for the therapy of renal cell carcinoma by acting as an inhibitor of mTOR and subsequently inducing the autophagy of cancer cells [133].

3.3. Natural Product-Based ADCs

Cancer immunotherapies, including chimeric antigen receptor T cells, immune checkpoint blocks and vaccines, have been developed as promising treatments and have made significant therapeutic progresses in recent years. In contrast to traditional therapies, cancer immunotherapies have high accuracy, specificity and wide adaptability and significantly improve patient survival rate [134]. Monoclonal antibodies target immune checkpoints, which has revolutionized cancer treatment. Monoclonal antibodies can be used alone as immune checkpoint inhibitors. Additionally, they can be conjugated with potent cytotoxic agents and then deliver the cytotoxic agents precisely to the tumor cells.
Trastuzumab emtansine is a human epidermal growth factor receptor 2-targeted monoclonal antibody conjugated to emtansine. Brentuximab vedotin is an anti-CD30 antibody conjugated to monomethyl auristatin E. Both of these drugs improve cancer therapy efficacy by inhibiting microtubule generation via disrupting the polymerization of tubulin [135,136]. Both inotuzumab ozogamicin and gemtuzumab ozogamicin consist of a monoclonal antibody and a cytotoxic calicheamicin that bind DNA and subsequently result in DNA breaks. The former is directly delivered to refractory or relapsed acute lymphoblastic leukemia cells by anti-22 antibody and then induces DNA damage [137]. The latter is directly delivered to acute myeloid leukemia cells by anti-CD33 antibody. In general, to a certain extent, ADCs reduce the toxicity of drugs to normal cells due to their specificity and exhibit greater potential in cancer treatment than traditional therapy. However, autoimmune response and side effects still exist, and treatment cost is high, all of which hinder their widespread usage. Future developments in ADCs will focus on identifying better targets, more effective cytotoxic payloads and better linkers to improve the potency and safety of drugs.

3.4. Marine Natural Products

Compared with terrestrial plants, nonmarine microorganisms and animals, marine organisms possess greater potential to produce novel and bioactive NPs with diverse structures. They are considered to be the most recent sources of medicinal drugs. As of 2021, four marine NPs have been approved for cancer therapy.
The first marine anticancer drug to be approved was eribulin, which binds tubulin and induces microtubule depolymerization in breast cancer and soft-tissue sarcoma [138]. In addition, there are two marine drugs targeting transcriptional regulation, trabectedin and lurbinectedin. Trabectedin is produced from the marine animal Ecteinascidia turbinate. It is used to treat specific soft tissue sarcomas, such as liposarcoma and leiomyosarcoma. According to a clinical study, treatment with trabectedin produces better efficacy and a longer survival time in soft-tissue sarcoma patients [139]. Aplidine is produced from the marine microorganism Aplidium albicans, and it has curative effects in acute lymphoblastic leukemia via oxidative stress-mediated JNK and p38 activation and ER stress-mediated incorrect protein folding, triggering rapid apoptosis in cancer cells [140]. Lurbinectedin is a synthetic alkaloid related to trabectedin. As a new second-line therapy option, it was approved in 2021 for the treatment of metastatic small cell lung cancer. Lurbinectedin acts as a transcription inhibitor by binding to the minor groove of DNA and subsequently driving the accumulation of DNA breaks. Additionally, it can disrupt the interactions between DNA and protein and thereby disrupt RNA transcription [141].
With the development of techniques for sample collection and spectrometry, significant achievements have been made in the exploration of marine natural products. More than 32,900 marine NPs had been identified by 2020 [142,143,144,145]. The quantity of marine NPs is relatively high compared to that of synthetic compounds. Thus, their clinical trials are very promising.

3.5. Hormones and Other Natural Products

Hormones and their analogues suppress cancer development mainly by regulating androgen or estrogen levels. For example, formestane and testolactone function as aromatase inhibitors and decrease the estrogen levels of breast cancer and unresectable desmoid tumors, respectively [146,147]. Abiraterone acetate decreases the androgen level by acting as an inhibitor of CYP17 [148]. However, only one hormone analogue has been approved for cancer therapy. It seems that hormones are becoming less widespread in clinical use.
Moreover, some other NPs can also inhibit cancer development by modulating the immune system. Purine analogues and peptide analogues, such as cladribine and mifamurtide, have been shown to be active in a variety of B- and T cell malignancies [149,150]. They can activate immune cells, inducing an effective immune response and resulting in protection against cancer. Some cancer cells, such as multiple myeloma cells, are sensitive to the inhibition of the ubiquitin proteasome pathway induced by carfilzomib, since proteasome inhibition leads to the accumulation of unfolded proteins, endoplasmic reticulum stress and subsequent cell growth arrest [151]. As a modified epoxyketone, carfilzomib can bind to the functional sites of the 20S proteasome, inhibiting cell proliferation [152].
Photodynamic therapy is another attractive treatment technology. Unlike standard laser photocoagulation, photodynamic therapy enhances the antitumor effects by inducing vascular shutdown [153,154]. Vascular shutdown serves as a promising target for anticancer agents that bind to tubulin protein and lead to microtubule depolymerization [155] (Figure 3C). Similarly, talaporfin sodium evokes vascular shutdown via the RhoA/ROCK pathway, which directly mediates F-actin polymerization and subsequently destroys tumor vessels [110,156] (Figure 3C). The trastuzumab emtansine-induced disruption of microtubule dynamics summarized above can also block the formation of proplatelets and blood vessels [135].

4. Synthetic Biology Strategies and Tools for Discovering Natural Products

Given the great potential of NPs in clinical applications, an increasing number of pharmaceutical researchers have focused on NPs. Traditional methods are the primary approaches for discovering NPs because they involve the direct collection of compounds with biological activities from numerous organisms instead of genetic manipulation. Although traditional methods have been successfully applied in identifying many bioactive NPs, they have some limitations, such as being time-consuming, labor-consuming, having poor efficiency and requiring repeated extraction of known compounds. With the rapid development of sequencing, many genome sequences, RNA sequences and protein sequences have been shared in databases, such as the National Center for Biotechnology Information (NCBI) databases and UniProt. All these sequences light the avenues of bioinformatics, genetic manipulation, protein engineering and subsequent synthetic biology.

4.1. Bioinformatics Analysis

Databases provide vast information for researchers. For example, PubMed contains more than 26 million biomedical publications, which cover life science, chemical science, biological engineering, physical science and so on [157]. Based on the literature, researchers can extract target information that provides new insights for the therapeutic discovery and improvement of NPs against cancers. In addition, a series of genome analysis, proteome analysis and pathway analysis tools have been developed to discover hidden information. For example, antiSMASH is updated frequently and is used to predict and analyze the biosynthetic gene clusters (BGCs) of secondary metabolites in bacterial, fungal and plant genome sequences [16,158,159,160,161] (Figure 4A). These tools are constantly being extended and improved to help researchers identify unique and valuable metabolites.
High-throughput sequencing technologies are widely applied in genomics, transcriptomics, proteomics, metabolomics, lipidomics and single-cell sequencing (Figure 4B). These omics studies fill the knowledge gap regarding how many silent BGCs will yield novel NPs and then break through the bottleneck in BGC characterization [162]. Multiomics analysis provides crucial clues for revealing new insights into the biosynthetic mechanisms of NPs by integrating multilayer molecular information [163,164]. On the other hand, with the development of multiomics technologies and computer programs, there are enough genome sequences, RNA sequences, protein sequences and metabolites to be used for computational modeling and the subsequent generation of predictive biological systems and even metabolic reconstructions [165]. In a computational metabolic model, researchers can obtain a simulation that resembles a real laboratory experiment and an approximate outcome of the experiment by running a model in which researchers can adjust the parameters of every factor in the biosynthetic pathway to optimize the output of the end product (Figure 4C). For example, Michael C et al. achieved a three-fold improvement in the resveratrol titer in Escherichia coli (E. coli) by constructing a new probabilistic computational model [166]. In addition to metabolic modeling, computational models are also used to design cell factories to realize the optimal production of target molecules [167].

4.2. Pathway Reconstruction or Engineering

However, 90% of the NPs are in the dark, as they are produced at only low levels or not at all due to their silent BGCs. The rapid development of DNA sequencing technology has stimulated genetic manipulation, which is crucial for identifying and engineering novel and valuable NPs. Multiple-step biosynthetic pathways are known to be involved in NP biosynthesis, and these pathways contain diverse genes and their control elements. Researchers will be better able to discover these high-hanging fruits if these genes and their elements are optimally assembled into operational pathway(s). Therefore, new DNA assembly and engineering tools are arising constantly.
Pathway construction through DNA assembly in vivo or in vitro remains a fast and efficient method [168] (Figure 5A). Type II restriction enzyme-directed assembly of multiple DNA fragments in vitro and its derived technologies have various advantages due to their short cycle and convenient operations. On the other hand, the assembly of multiple overlapping DNA fragments based on homologous recombination in vitro or in vivo, as with Gibson Assembly, In-Fusion Snap Assembly and other assembly kits, is also popular in the laboratory because it can achieve seamless assembly. It can even realize a seamless construct of an entire bacterial genome (total size of 583 kb) or even a 900 kb DNA product with high efficiency [169]. However, errors may be introduced during in vitro recombination. Thus, homologous recombination in vivo, such as in yeast and E. coli, is powerful. It has not only high efficiency but also a low error rate. Additionally, pathway construction by direct cloning has gradually developed and matured (Figure 5B). The classic and widely used direct cloning method is genome library construction, which is completed by digesting the genome using restriction endonucleases and then ligating the genome fragments into expression vectors using T4. Bacterial artificial chromosome (BAC) libraries and transformation-associated recombination (TAR) are commonly used for NP discovery in the laboratory [170]. However, these methods are aimless, time-consuming and labor-intensive. Cas9-assisted targeting of chromosome segments (CATCH), a cloning technology that is designed based on CIRSPR-Cas9-mediated targeting digestion and Gibson assembly-mediated ligation, makes up for the shortcomings of seamless assembly [171]. This approach can be used to clone target genomic DNA sequences with a size of up to 100 kb. Another technology, RecET direct cloning, bypasses library construction and screening and directly clones large genomic DNA fragments into expression vector via RecET-mediated homologous recombination and Redαβ-mediated recombination [20,172].
Pathway engineering via single base or module editing based on CRISPR systems is a powerful strategy for discovering NPs and their derivatives and for improving NP titers (Figure 5C). Limited enzyme activity and poor protein stability are bottlenecks in the biosynthesis of NPs. Thus, researchers have focused on random or site-directed mutagenesis to improve enzyme activity and protein stability. Chen H et al. increased the titer of lycopene in Saccharomyces cerevisiae by 2.53 times by improving the catalytic activity of isopentenyl diphosphate isomerase via random mutation [173]. CRISPR/Cas-mediated base editing, an efficient technology for precise single-base editing of the genome in vivo, can precisely convert a C:G base pair to a T:A base pair by ligating a cytidine deaminase to a dead Cas (dCas) protein or convert an A:T base pair to a G:C base pair by ligating an adenosine deaminase to a dCas protein [174,175,176]. Gene deletion or insertion is another highly efficient method for NP discovery and titer improvement. The deletion of some negative regulators, competitive pathways or negative feedback pathways can lead to significant accumulation of an NP [177,178,179]. On the other hand, increasing the expression levels of key synthetic enzymes, rate-limiting enzymes or positive transcription regulators can also prominently improve NP production [180,181,182]. The production of spinosad showed a 1000-fold increase compared to its original production after overexpressing rate-limiting proteins by introducing strong promoters upstream of rate-limiting genes [183]. Among the NP biosynthetic enzymes, the sequences in modular NRPS or PKS show high similarities. Thus, engineering NRPS or PKS modular enzymes, including repeating the target functional modules and replacing a functional module with another functional or replacing the module with a heterologous module, will provide a platform to generate almost any desired derivative. Kudo K et al. obtained 19 rapamycin derivatives by editing modular polyketide synthase genes of rapamycin. CRISPR-Cas9, CRISPR-Cas12a and their derived tools are widely used to accurately edit single nucleotides, modules, genes or gene clusters [183].

4.3. Cell Factory

Host organisms are critical in the heterologous production of NPs because their metabolic efflux greatly influences NP production directly. Generally, natural producers are imperfect hosts for the production of NPs due to the lack of sufficient precursors or efficient genetic engineering techniques, slow growth rates or environmental perturbations. Thus, heterologous hosts are needed to ensure the highly efficient expression of NPs.
The cell factory, a self-replicating minimal cell, provides an optimal metabolic background for the biosynthesis of NPs. In the cell factory, genome mining is generally performed (Figure 6A). The nonessential genes or gene clusters are deleted from the genome to reduce the number of competing pathways; at the same time, extra integration sites are inserted into the genome to allow additional integration of multiple copies of a heterologous gene cluster [184,185]. Additionally, genetic regulation is an effective approach to optimize the cell factory. A series of regulators have been characterized and proven to be functional in NP biosynthesis [177,186]. Moreover, promoter engineering [187], transcription factor engineering [188], synthetic RNA switch and CRISPR-Cas systems [189] are also widely used to regulate gene expression and subsequent NP production in cell factories. Protein engineering has also become an increasingly popular method to improve protein activity, increase protein stability, maximize carbon flux through limiting steps, expand NP spectra and improve NP production [22]. A more complex issue is that the cell factory should be able to synthesize its own essential nutrients instead of acquiring them from the environment [190]. Therefore, understanding the minimum requirements and designing the cell factory from scratch are necessary. Metabolism engineering, including primary and secondary metabolism engineering, significantly increases the product pathway flux of target products due to the accumulation or balance of metabolic precursors, cofactors and energy. Metabolism engineering provides an optimal metabolic background for heterologous expression.
Microorganisms are the most popular cell factories, as they are easy to culture and genetically manipulate. E. coli and Saccharomyces cerevisiae (S. cerevisiae) are widely used due to their fast growth rates, easy genetic manipulations and high productivities [24]. Moreover, as the second family in which NPs were discovered, the Streptomycetaceae family has attracted much attention for its prolific drugs [191]. Streptomyces strains, especially Streptomyces coelicolor and Streptomyces albus [184], have rich cofactors and precursors. They are widely reengineered to construct efficient cell factories. For example, Myronovskyi M et al. constructed a cluster-free Streptomyces albus chassis strain by deleting these unessential gene clusters and introducing an additional phage phiC31 attB site to increase the copy number of heterologous gene clusters. In the last, the authors significantly improved the production of six compounds including pyridinopyrone A, aloesaponarin II and didesmethyl-mensacarcin [184]. Moreover, E. coli and yeast strains have already been metabolically redesigned and engineered to overproduce intermediates of paclitaxel [192,193,194]. Although microorganisms exhibit many advantages, they have some limitations. For example, not all plant NPs, animal NPs and marine NPs can be expressed in microorganism hosts, as they lack efficient biosynthetic pathways or enzymes. To overcome these limitations, more and more plant cell factories [195], cyanobacteria cell factories and other cell factories have been developed. These heterologous plant or marine hosts provide alternative and sustainable avenues for the production of NPs that are not suitable for expression in microbial hosts.

4.4. Artificial Intelligence

With the rapid development of artificial intelligence, robotics and integrated software are also widely used to automate standard workflows, in which researchers write programs and robots complete the experiments according to the programs (Figure 6B). This workflow significantly improves the efficiency of synthetic biology and subsequently improves the speed of researchers to explore and create active compounds. Synthetic biology is a type of “on-demand manufacturing” composed of a variety of nonlinear workflows. However, the heterogeneity and dynamics of biological experiments make the workflows more complex due to the differences among batches [196]. In recent years, biological foundries have been developed with the integration of the automatic design–build–test engineering cycle, which solves several difficult problems in the processes of bioengineering, such as slow speed, high expense and low repeatability [197]. Mohammad H. et al. reported an application of an integrated robot system combined with a machine learning algorithm; this application completes the design, construction, testing and learning process of a fully automated biological system [198]. The fully automated robotic platform IOAutomata evaluated slightly less than 1% of the possible variation, which was 77% higher than that achieved by random screening. The authors also successfully used the robotic platform to optimize the biosynthesis of lycopene.

5. Conclusions and Future Perspectives

NPs have great potential in clinical cancer treatment due to their diverse structures and bioactivities. An increasing number of NPs, especially marine drugs and ADCs, have been applied in cancer treatment. With the emerging demand for novel natural drugs, effective methods for discovering NPs are needed. Although traditional approaches have been successfully applied to identify a large number of bioactive NPs, these approaches remain limited, as they are time-consuming, labor-intensive, “low-hanging” and inconsistent. With the conspicuous progress in genetic manipulation approaches and detection technologies, NP discovery and production improvement have become more promising.
Synthetic biology greatly promotes the exploitation of NPs. From traditional sample collection to complex genetic manipulation, and then to artificial intelligence, the exploitation rates and quantity of NPs are gradually increasing. For example, complex and powerful genetic manipulation greatly optimizes biosynthetic pathways and metabolic networks and provides an optimal background for target NP biosynthesis. Moreover, with synthetic biology, known compounds can also be modified through targeted design to obtain analogues with better activity and less toxicity or even new compounds. Specifically, through the application of artificial intelligence, more and more fully automated and multifunctional robotic platforms have been built and integrated. These systems overcome the above limits in the processes of bioengineering and greatly improve their efficiency and accuracy. Thus, efforts in synthetic biology will help NP discovery continue to thrive in the future.
Among the approved anticancer NPs, marine and antibody-conjugated NPs have rapidly emerged in the last decade (Figure 1D). These 32,900 marine natural products are only the tip of the iceberg. The organisms in the deep oceans have undergone the longest evolutionary period, and the harsh marine conditions have endowed them with a wide range of unique molecules. Thus, the potential of marine NPs in future drug discovery and clinical cancer treatment is immeasurable.
Additionally, smart drug delivery systems show strong competitiveness in cancer treatment. Smart delivery can improve the permeability, stability and solubility of drugs, promote the continuous controlled release of drugs and increase the therapeutic effects. In these smart delivery systems, the NPs are precisely delivered to target cancer cells, resulting in high toxicity to cancer and reduced side effects on the human body [199]. Although various drug delivery strategies have been explored to deliver chemotherapeutic drugs more directly to tumors, the most important transformation progresses have appeared in the field of ADCs. As an immune therapy, NP-derived ADCs represent novel biological systems for target treatment. Moreover, some NPs directly interact with and activate immune cells or pathways, often inducing protection against cancer [15]. Therefore, NPs also realize their clinical significance via immunotherapy in cancer treatment.
Furthermore, the complexity of refractory diseases has greatly weakened the therapeutic potential of existing treatment schemes. Therefore, clinical treatment has changed from monotherapy to combination therapy, which is now becoming accepted as an effective way to optimize efficacy while minimizing adverse effects [200]. Polatuzumab vedotin was first approved for the treatment of hematological malignancies, and combination therapy with rituximab produces a more significant efficacy than monotherapy in diffuse large B-cell lymphoma [201]. In addition to chemotherapy, polatuzumab vedotin can also be used with immunomodulatory therapy, which is promising for relapsed/refractory follicular lymphoma [202].
In summary, considering their various structures and bioactivities, the potential of NPs in clinical cancer treatment is highly promising. Specifically, emerging marine NPs and ADCs provide more choices of medications with higher efficacy and safety. Furthermore, combination therapy not only increases therapeutic efficacy but also helps to address drug resistance [203]. With the innovation of various technologies and popularization of artificial intelligence, a large number of NPs are being brought to our sights, providing more candidate clinical medications.

Author Contributions

Y.H. (Yu He) and D.Z. drafted the manuscript; L.L., H.W., Y.H. (Yujia He), M.J., X.F., Y.C., J.F. and D.Z. reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of China (No. 82121003), Science & Technology Department of Sichuan Province (2021JDZH0031, 2022YFS0369), Clinical Research and Transformation Fund of Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital (2021LY04) and Health Care Committee of Sichuan Province (CGY2021-227).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare that there are no conflict of interest.

Abbreviations

ADCAntibody drug conjugate
AntiSMASHAntibiotics and secondary metabolite analysis shell
BACBacterial artificial chromosome
BGCBiosynthetic gene cluster
CATCHCas9-assisted targeting of chromosome segment
CRISPR-Casclustered regularly interspaced short palindromic repeats-CRISPR associated protein
DCasDead Cas
DSBDouble-strand break
E. coliEscherichia coli
sNCBINational center for biotechnology information
NPNatural product
NRPSNonribosomal peptide synthetase
PKSPolyketide synthase
RiPPsRibosomally synthesized and posttranslationally modified peptides
S. cerevisiaeSaccharomyces cerevisiae
TARTransformation-associated recombination
U.S. FDAUnited States Food and Drug Administration

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2020, 70, 209–249. [Google Scholar] [CrossRef]
  2. Parkin, D.M.; Bray, M.F.; Ferlay, M.J.; Pisani, P. Global Cancer Statistics, 2002. CA Cancer J. Clin. 2005, 55, 74. [Google Scholar] [CrossRef] [PubMed]
  3. Shiri, P.; Ramezanpour, S.; Amani, A.M.; Dehaen, W. A patent review on efficient strategies for the total synthesis of pazopanib, regorafenib and lenvatinib as novel anti-angiogenesis receptor tyrosine kinase inhibitors for cancer therapy. Mol. Divers. 2022, 26, 2981–3002. [Google Scholar] [CrossRef] [PubMed]
  4. Mashayekh, K.; Shiri, P. An overview of recent advances in the applications of click chemistry in the synthesis of bioconjugates with anticancer activities. ChemistrySelect 2019, 4, 13459–13478. [Google Scholar] [CrossRef]
  5. Tsai, C.H.; Lin, Y.H.; Li, Y.S.; Ho, T.L.; Hoai Thuong, L.H.; Liu, Y.H. Integrated medicine for chemotherapy-induced peripheral neuropathy. Int. J. Mol. Sci. 2021, 22, 9257. [Google Scholar] [CrossRef] [PubMed]
  6. Daudon, M.; Frochot, V.; Bazin, D.; Jungers, P. Drug-induced kidney stones and crystalline nephropathy: Pathophysiology, prevention and treatment. Drugs 2018, 78, 163–201. [Google Scholar] [CrossRef]
  7. Katz, L.; Baltz, R.H. Natural product discovery: Past, present, and future. J. Ind. Microbiol. Biot. 2016, 43, 155–176. [Google Scholar] [CrossRef]
  8. Ashrafizadeh, M.; Mirzaei, S.; Hashemi, F.; Zarrabi, A.; Zabolian, A.; Saleki, H.; Sharifzadeh, S.O.; Soleymani, L.; Daneshi, S.; Hushmandi, K.; et al. New insight towards development of paclitaxel and docetaxel resistance in cancer cells: EMT as a novel molecular mechanism and therapeutic possibilities. Biomed. Pharmacother. 2021, 141, 111824. [Google Scholar] [CrossRef]
  9. Liu, Y.; Yang, F.; Zou, S.; Qu, L. Rapamycin: A bacteria-derived immunosuppressant that has anti-atherosclerotic effects and its clinical application. Front. Pharmacol. 2018, 9, 1520. [Google Scholar] [CrossRef]
  10. Talman, A.M.; Clain, J.; Duval, R.; Menard, R.; Ariey, F. Artemisinin bioactivity and resistance in malaria parasites. Trends Parasitol. 2019, 35, 953–963. [Google Scholar] [CrossRef]
  11. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Xu, M.; Wright, G.D. Heterologous expression-facilitated natural products’ discovery in actinomycetes. J. Ind. Microbiol. Biot. 2019, 46, 415–431. [Google Scholar] [CrossRef] [PubMed]
  13. Ma, W.; Zhang, D.; Li, G.; Liu, J.; He, G.; Zhang, P.; Yang, L.; Zhu, H.; Xu, N.; Liang, S. Antibacterial mechanism of daptomycin antibiotic against Staphylococcus aureus based on a quantitative bacterial proteome analysis. J. Proteom. 2017, 150, 242–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Huang, X.M.; Yang, Z.J.; Xie, Q.; Zhang, Z.K.; Zhang, H.; Ma, J.Y. Natural products for treating colorectal cancer: A mechanistic review. Biomed. Pharmacother. 2019, 117, 109142. [Google Scholar] [CrossRef] [PubMed]
  15. Ri, M.H.; Ma, J.; Jin, X. Development of natural products for anti-PD-1/PD-L1 immunotherapy against cancer. J. Ethnopharmacol. 2021, 281, 114370. [Google Scholar] [CrossRef] [PubMed]
  16. Blin, K.; Shaw, S.; Steinke, K.; Villebro, R.; Ziemert, N.; Lee, S.Y.; Medema, M.H.; Weber, T. AntiSMASH 5.0: Updates to the secondary metabolite genome mining pipeline. Nucleic Acids Res. 2019, 47, W81–W87. [Google Scholar] [CrossRef] [Green Version]
  17. Goh, Y.J.; Barrangou, R. Harnessing CRISPR-Cas systems for precision engineering of designer probiotic lactobacilli. Curr. Opin. Biotechnol. 2018, 56, 163–171. [Google Scholar] [CrossRef]
  18. Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; Joung, J.; van der Oost, J.; Regev, A.; et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 2015, 163, 759–771. [Google Scholar] [CrossRef] [Green Version]
  19. Shao, Z.; Zhao, H.; Zhao, H. DNA assembler, an in vivo genetic method for rapid construction of biochemical pathways. Nucleic Acids Res. 2009, 37, e16. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, H.; Li, Z.; Jia, R.; Yin, J.; Li, A.; Xia, L.; Yin, Y.; Muller, R.; Fu, J.; Stewart, A.F.; et al. ExoCET: Exonuclease in vitro assembly combined with RecET recombination for highly efficient direct DNA cloning from complex genomes. Nucleic Acids Res. 2018, 46, e28. [Google Scholar] [CrossRef]
  21. Mao, X.M.; Luo, S.; Zhou, R.C.; Wang, F.; Yu, P.; Sun, N.; Chen, X.X.; Tang, Y.; Li, Y.Q. Transcriptional regulation of the daptomycin gene cluster in Streptomyces roseosporus by an autoregulator, AtrA. J. Biol. Chem. 2015, 290, 7992–8001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, C.; Zhang, R.; Wang, J.; Wilson, L.M.; Yan, Y. Protein engineering for improving and diversifying natural product biosynthesis. Trends Biotechnol. 2020, 38, 729–744. [Google Scholar] [CrossRef]
  23. Chandra Mohana, N.; Yashavantha Rao, H.C.; Rakshith, D.; Mithun, P.R.; Nuthan, B.R.; Satish, S. Omics based approach for biodiscovery of microbial natural products in antibiotic resistance era. J. Genet. Eng. Biotechnol. 2018, 16, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, X.; Liu, Y.; Du, G.; Ledesma-Amaro, R.; Liu, L. Microbial chassis development for natural product biosynthesis. Trends Biotechnol. 2020, 38, 779–796. [Google Scholar] [CrossRef] [PubMed]
  25. Yuan, R.K.; Hou, Y.; Sun, W.; Yu, J.; Liu, X.; Niu, Y.A.; Lu, J.J.; Chen, X.P. Natural products to prevent drug resistance in cancer chemotherapy: A review. Ann. N. Y. Acad. Sci. 2017, 1401, 19–27. [Google Scholar] [CrossRef] [PubMed]
  26. Man, S.; Luo, C.; Yan, M.; Zhao, G.; Ma, L.; Gao, W. Treatment for liver cancer: From sorafenib to natural products. Eur. J. Med. Chem. 2021, 224, 113690. [Google Scholar] [CrossRef]
  27. Kumar, A.; Jaitak, V. Natural products as multidrug resistance modulators in cancer. Eur. J. Med. Chem. 2019, 176, 268–291. [Google Scholar] [CrossRef]
  28. Khalifa, S.A.M.; Elias, N.; Farag, M.A.; Chen, L.; Saeed, A.; Hegazy, M.F.; Moustafa, M.S.; Abd El-Wahed, A.; Al-Mousawi, S.M.; Musharraf, S.G.; et al. Marine natural products: A source of novel anticancer drugs. Mar. Drugs 2019, 17, 491. [Google Scholar] [CrossRef] [Green Version]
  29. Dutta, S.; Mahalanobish, S.; Saha, S.; Ghosh, S.; Sil, P.C. Natural products: An upcoming therapeutic approach to cancer. Food Chem. Toxicol. 2019, 128, 240–255. [Google Scholar] [CrossRef]
  30. Cai, J.; Yi, M.; Tan, Y.; Li, X.; Li, G.; Zeng, Z.; Xiong, W.; Xiang, B. Natural product triptolide induces GSDME-mediated pyroptosis in head and neck cancer through suppressing mitochondrial hexokinase-II. J. Exp. Clin. Cancer Res. 2021, 40, 190. [Google Scholar] [CrossRef]
  31. Salzer, W.L.; Asselin, B.L.; Plourde, P.V.; Corn, T.; Hunger, S.P. Development of asparaginase Erwinia chrysanthemi for the treatment of acute lymphoblastic leukemia. Ann. N. Y. Acad. Sci. 2014, 1329, 81–92. [Google Scholar] [CrossRef]
  32. Nambiar, S.; Madurawe, R.D.; Zuk, S.M.; Khan, S.R.; Ellison, C.D.; Faustino, P.J.; Mans, D.J.; Trehy, M.L.; Hadwiger, M.E.; Boyne, M.T., 2nd; et al. Product quality of parenteral vancomycin products in the United States. Antimicrob. Agents Chemother. 2012, 56, 2819–2823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Jain, M.M.; Gupte, S.U.; Patil, S.G.; Pathak, A.B.; Deshmukh, C.D.; Bhatt, N.; Haritha, C.; Govind Babu, K.; Bondarde, S.A.; Digumarti, R.; et al. Paclitaxel injection concentrate for nanodispersion versus nab-paclitaxel in women with metastatic breast cancer: A multicenter, randomized, comparative phase II/III study. Breast Cancer Res. Treat. 2016, 156, 125–134. [Google Scholar] [CrossRef] [Green Version]
  34. Liu, H.Y.; Dong, T.X.; Li, Z.Z.; Li, T.T.; Jiang, J.; Zhu, M.W.; An, T.T.; Chen, Y.D.; Yang, X.H. Homoharringtonine inhibits the progression of hepatocellular carcinoma by suppressing the PI3K/AKT/GSK3β/Slug signaling pathway. Neoplasma 2021, 68, 924–937. [Google Scholar] [CrossRef] [PubMed]
  35. Hammadi, R.; Kúsz, N.; Dávid, C.Z.; Behány, Z.; Papp, L.; Kemény, L.; Hohmann, J.; Lakatos, L.; Vasas, A. Ingol and ingenol-type diterpenes from euphorbia trigona miller with keratinocyte inhibitory activity. Plants 2021, 10, 1206. [Google Scholar] [CrossRef] [PubMed]
  36. Chand, P.; Kumar, H.; Badduri, N.; Gupta, N.V.; Bettada, V.G.; Madhunapantula, S.V.; Kesharwani, S.S.; Dey, S.; Jain, V. Design and evaluation of cabazitaxel loaded NLCs against breast cancer cell lines. Colloids Surf. B. Biointerfaces 2021, 199, 111535. [Google Scholar] [CrossRef] [PubMed]
  37. Bianconi, M.; Cimadamore, A.; Faloppi, L.; Scartozzi, M.; Santoni, M.; Lopez-Beltran, A.; Cheng, L.; Scarpelli, M.; Montironi, R. Contemporary best practice in the management of urothelial carcinomas of the renal pelvis and ureter. Ther. Adv. Urol. 2019, 11, 1756287218815372. [Google Scholar] [CrossRef]
  38. Escrich, A.; Almagro, L.; Moyano, E.; Cusido, R.M.; Bonfill, M.; Hosseini, B.; Palazon, J. Improved biotechnological production of paclitaxel in Taxus media cell cultures by the combined action of coronatine and calix[8]arenes. Plant Physiol. Biochem. 2021, 163, 68–75. [Google Scholar] [CrossRef]
  39. Kawashiri, T.; Inoue, M.; Mori, K.; Kobayashi, D.; Mine, K.; Ushio, S.; Kudamatsu, H.; Uchida, M.; Egashira, N.; Shimazoe, T. Preclinical and clinical evidence of therapeutic agents for paclitaxel-induced peripheral neuropathy. Int. J. Mol. Sci. 2021, 22, 8733. [Google Scholar] [CrossRef]
  40. Yassine, F.; Salibi, E.; Gali-Muhtasib, H. Overview of the formulations and analogs in the taxanes’ story. Curr. Med. Chem. 2016, 23, 4540–4558. [Google Scholar] [CrossRef]
  41. Hur, J.; Ghosh, M.; Kim, T.H.; Park, N.; Pandey, K.; Cho, Y.B.; Hong, S.D.; Katuwal, N.B.; Kang, M.; An, H.J.; et al. Synergism of AZD6738, an ATR inhibitor, in combination with belotecan, a camptothecin analogue, in chemotherapy-resistant ovarian cancer. Int. J. Mol. Sci. 2021, 22, 1223. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, X.; Song, L.; Li, N.; Qiu, Z.; Zhou, S.; Li, C.; Zhao, J.; Song, H.; Chen, X. Pharmacokinetics and biodistribution study of paclitaxel liposome in Sprague-Dawley rats and Beagle dogs by liquid chromatography-tandem mass spectrometry. Drug. Res. 2013, 63, 603–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Curran, M.; Wiseman, L. Fulvestrant. Drugs 2001, 61, 807–813; discussion 814. [Google Scholar] [CrossRef] [PubMed]
  44. Lone, S.H.; Bhat, K.A.; Khuroo, M.A. Arglabin: From isolation to antitumor evaluation. Chem. Biol. Interact. 2015, 240, 180–198. [Google Scholar] [CrossRef] [PubMed]
  45. Riemsma, R.; Simons, J.P.; Bashir, Z.; Gooch, C.L.; Kleijnen, J. Systematic review of topotecan (Hycamtin) in relapsed small cell lung cancer. BMC Cancer 2010, 10, 436. [Google Scholar] [CrossRef] [Green Version]
  46. Hande, K.R. Etoposide: Four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 1998, 34, 1514–1521. [Google Scholar] [CrossRef]
  47. Sharma, S.; Pukale, S.S.; Sahel, D.K.; Agarwal, D.S.; Dalela, M.; Mohanty, S.; Sakhuja, R.; Mittal, A.; Chitkara, D. Folate-targeted cholesterol-grafted lipo-polymeric nanoparticles for chemotherapeutic agent delivery. AAPS Pharm. Sci. Technol. 2020, 21, 280. [Google Scholar] [CrossRef]
  48. Ando, K.; Ozonoff, A.; Lee, S.Y.; Voisine, M.; Parker, J.T.; Nakanishi, R.; Nishimura, S.; Yang, J.; Grace, Z.; Tran, B.; et al. Multicohort retrospective validation of a predictive biomarker for topoisomerase I inhibitors. Clin. Colorectal. Cancer 2021, 20, e129–e138. [Google Scholar] [CrossRef]
  49. Yang, C.H.; Horwitz, S.B. Taxol(®): The first microtubule dtabilizing agent. Int. J. Mol. Sci. 2017, 18, 1733. [Google Scholar] [CrossRef] [Green Version]
  50. Epstein, E. Warning! Masoprocol is a potent sensitizer. J. Am. Acad. Dermatol. 1994, 31, 295–297. [Google Scholar] [CrossRef]
  51. Reinhorn, D.; Kuchuk, I.; Shochat, T.; Nisenbaum, B.; Sulkes, A.; Hendler, D.; Rotem, O.; Tsoref, D.; Olitzky, O.; Goldvaser, H.; et al. Taxane versus vinorelbine in combination with trastuzumab and pertuzumab for first-line treatment of metastatic HER2-positive breast cancer: A retrospective two-center study. Breast Cancer Res. Treat. 2021, 188, 379–387. [Google Scholar] [CrossRef] [PubMed]
  52. Wu, J.; Tang, X.; Ma, C.; Shi, Y.; Wu, W.; Hann, S.S. The regulation and interaction of colon cancer-associated transcript-1 and miR7-5p contribute to the inhibition of SP1 expression by solamargine in human nasopharyngeal carcinoma cells. Phytother. Res. 2020, 34, 201–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hiyani, L.E.; Samperez, S.; Jouan, P. Inhibition by Celiptium of the fetal thymidine kinase synthesis induced by estrogens in the rat uterus. Chem. Biol. Interact. 1987, 62, 167–178. [Google Scholar] [CrossRef]
  54. Zhang, W.; Gou, P.; Dupret, J.M.; Chomienne, C.; Rodrigues-Lima, F. Etoposide, an anticancer drug involved in therapy-related secondary leukemia: Enzymes at play. Transl. Oncol. 2021, 14, 101169. [Google Scholar] [CrossRef] [PubMed]
  55. Mondal, A.; Gandhi, A.; Fimognari, C.; Atanasov, A.G.; Bishayee, A. Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur. J. Pharmacol. 2019, 858, 172472. [Google Scholar] [CrossRef] [PubMed]
  56. Zalesak, F.; Bon, D.J.D.; Pospisil, J. Lignans and neolignans: Plant secondary metabolites as a reservoir of biologically active substances. Pharmacol. Res. 2019, 146, 104284. [Google Scholar] [CrossRef]
  57. Vellemans, H.; André, M.P.E. Review of treatment options for the management of advanced stage hodgkin lymphoma. Cancers 2021, 13, 3745. [Google Scholar] [CrossRef]
  58. Zhao, J.C.; Agarwal, S.; Ahmad, H.; Amin, K.; Bewersdorf, J.P.; Zeidan, A.M. A review of FLT3 inhibitors in acute myeloid leukemia. Blood Rev. 2022, 52, 100905. [Google Scholar] [CrossRef]
  59. Romidepsin. In LiverTox: Clinical and Research Information on Drug-Induced Liver Injury; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2012.
  60. Bukowski, R.M. Temsirolimus: A safety and efficacy review. Expert. Opin. Drug Saf. 2012, 11, 861–879. [Google Scholar] [CrossRef]
  61. Edelman, M.J.; Shvartsbeyn, M. Epothilones in development for non--small-cell lung cancer: Novel anti-tubulin agents with the potential to overcome taxane resistance. Clin. Lung Cancer 2012, 13, 171–180. [Google Scholar] [CrossRef]
  62. Horita, N.; Yamamoto, M.; Sato, T.; Tsukahara, T.; Nagakura, H.; Tashiro, K.; Shibata, Y.; Watanabe, H.; Nagai, K.; Nakashima, K.; et al. Amrubicin for relapsed small-cell lung cancer: A systematic review and meta-analysis of 803 patients. Sci. Rep. 2016, 6, 18999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Onrust, S.V.; Lamb, H.M. Valrubicin. Drugs Aging 1999, 15, 69–75; discussion 76. [Google Scholar] [CrossRef] [PubMed]
  64. Okusaka, T.; Okada, S.; Ishii, H.; Ikeda, M.; Nakasuka, H.; Nagahama, H.; Iwata, R.; Furukawa, H.; Takayasu, K.; Nakanishi, Y.; et al. Transarterial chemotherapy with zinostatin stimalamer for hepatocellular carcinoma. Oncology 1998, 55, 276–283. [Google Scholar] [CrossRef]
  65. Ke, B.; Li, A.; Fu, H.; Kong, C.; Liu, T.; Zhu, Q.; Zhang, Y.; Zhang, Z.; Chen, C.; Jin, C. PARP-1 inhibitors enhance the chemosensitivity of leukemia cells by attenuating NF-кB pathway activity and DNA damage response induced by Idarubicin. Acta Biochim. Biophys. Sin. 2022, 54, 91–98. [Google Scholar] [CrossRef] [PubMed]
  66. Li, Q.; Qin, M.; Tan, Q.; Li, T.; Gu, Z.; Huang, P.; Ren, L. MicroRNA-129-1-3p protects cardiomyocytes from pirarubicin-induced apoptosis by down-regulating the GRIN2D-mediated Ca(2+) signalling pathway. J. Cell Mol. Med. 2020, 24, 2260–2271. [Google Scholar] [CrossRef] [Green Version]
  67. PLoSker, G.L.; Faulds, D. Epirubicin. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in cancer chemotherapy. Drugs 1993, 45, 788–856. [Google Scholar] [CrossRef]
  68. Wander, D.P.A.; van der Zanden, S.Y.; van der Marel, G.A.; Overkleeft, H.S.; Neefjes, J.; Codée, J.D.C. Doxorubicin and aclarubicin: Shuffling anthracycline glycans for improved anticancer agents. J. Med. Chem. 2020, 63, 12814–12829. [Google Scholar] [CrossRef] [PubMed]
  69. Carter, S.K.; Ultmann, J. Peplomycin. Cancer Treat. Rev. 1984, 11, 303–305. [Google Scholar] [CrossRef]
  70. Sankaran, H.; Sengupta, S.; Purohit, V.; Kotagere, A.; Moulik, N.R.; Prasad, M.; Dhamne, C.; Narula, G.; Banavali, S.; Gota, V. A comparison of asparaginase activity in generic formulations of E.coli derived L- asparaginase: In-vitro study and retrospective analysis of asparaginase monitoring in pediatric patients with leukemia. Br. J. Clin. Pharmacol. 2020, 86, 1081–1088. [Google Scholar] [CrossRef]
  71. Sinha, R.; Purkayastha, P. Daunomycin delivery by ultrasmall graphene quantum dots to DNA duplexes: Understanding the dynamics by resonance energy transfer. J. Mater. Chem. B 2020, 8, 9756–9763. [Google Scholar] [CrossRef]
  72. Bleomycin. Drugs and Lactation Database (LactMed); National Library of Medicine (US): Bethesda, MD, USA, 2006.
  73. Carvalho, C.; Santos, R.X.; Cardoso, S.; Correia, S.; Oliveira, P.J.; Santos, M.S.; Moreira, P.I. Doxorubicin: The good, the bad and the ugly effect. Curr. Med. Chem. 2009, 16, 3267–3285. [Google Scholar] [CrossRef]
  74. Dactinomycin. In LiverTox: Clinical and Research Information on Drug-Induced Liver Injury; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2012.
  75. Kormanec, J.; Novakova, R.; Csolleiova, D.; Feckova, L.; Rezuchova, B.; Sevcikova, B.; Homerova, D. The antitumor antibiotic mithramycin: New advanced approaches in modification and production. Appl. Microbiol. Biotechnol. 2020, 104, 7701–7721. [Google Scholar] [CrossRef] [PubMed]
  76. Sinawe, H.; Casadesus, D. Mitomycin. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  77. Bleyer, W.A. New vistas for leucovorin in cancer chemotherapy. Cancer 1989, 63, 995–1007. [Google Scholar] [CrossRef]
  78. Targeting nectin-4 in bladder cancer. Cancer Discov. 2017, 7, Of3. [CrossRef] [Green Version]
  79. Wang, Y.; Liu, L.; Fan, S.; Xiao, D.; Xie, F.; Li, W.; Zhong, W.; Zhou, X. Antibody-drug conjugate using ionized Cys-linker-MMAE as the potent payload shows optimal therapeutic safety. Cancers 2020, 12, 744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [Green Version]
  81. Kantarjian, H.M.; DeAngelo, D.J.; Stelljes, M.; Martinelli, G.; Liedtke, M.; Stock, W.; Gökbuget, N.; O’Brien, S.; Wang, K.; Wang, T.; et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N. Engl. J. Med. 2016, 375, 740–753. [Google Scholar] [CrossRef]
  82. Advani, R.H.; Moskowitz, A.J.; Bartlett, N.L.; Vose, J.M.; Ramchandren, R.; Feldman, T.A.; LaCasce, A.S.; Christian, B.A.; Ansell, S.M.; Moskowitz, C.H.; et al. Brentuximab vedotin in combination with nivolumab in relapsed or refractory Hodgkin lymphoma: 3-year study results. Blood 2021, 138, 427–438. [Google Scholar] [CrossRef]
  83. Lambert, J.; Pautas, C.; Terré, C.; Raffoux, E.; Turlure, P.; Caillot, D.; Legrand, O.; Thomas, X.; Gardin, C.; Gogat-Marchant, K.; et al. Gemtuzumab ozogamicin for de novo acute myeloid leukemia: Final efficacy and safety updates from the open-label, phase III ALFA-0701 trial. Haematologica 2019, 104, 113–119. [Google Scholar] [CrossRef] [Green Version]
  84. Baena, J.; Modrego, A.; Zeaiter, A.; Kahatt, C.; Alfaro, V.; Jimenez-Aguilar, E.; Mazarico, J.M.; Paz-Ares, L. Lurbinectedin in the treatment of relapsed small cell lung cancer. Future Oncol. 2021, 17, 2279–2289. [Google Scholar] [CrossRef]
  85. Le Tourneau, C.; Raymond, E.; Faivre, S. Aplidine: A paradigm of how to handle the activity and toxicity of a novel marine anticancer poison. Curr. Pharm. Des. 2007, 13, 3427–3439. [Google Scholar] [CrossRef] [PubMed]
  86. Cuevas, C.; Francesch, A. Development of Yondelis (trabectedin, ET-743). A semisynthetic process solves the supply problem. Nat. Prod. Rep. 2009, 26, 322–337. [Google Scholar] [CrossRef] [PubMed]
  87. Perry, C.M. Eribulin. Drugs 2011, 71, 1321–1331. [Google Scholar] [CrossRef] [PubMed]
  88. Abiraterone. In LiverTox: Clinical and Research Information on Drug-Induced Liver Injury; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2012.
  89. Calès, P. Vapreotide acetate for the treatment of esophageal variceal bleeding. Expert Rev. Gastroenter. Hepatol. 2008, 2, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Scott, L.J.; Wiseman, L.R. Exemestane. Drugs 1999, 58, 675–680; discussion 681–682. [Google Scholar] [CrossRef] [PubMed]
  91. Khoshghamat, N.; Jafari, N.; Toloue-Pouya, V.; Azami, S.; Mirnourbakhsh, S.H.; Khazaei, M.; Ferns, G.A.; Rajabian, M.; Avan, A. The therapeutic potential of renin-angiotensin system inhibitors in the treatment of pancreatic cancer. Life Sci. 2021, 270, 119118. [Google Scholar] [CrossRef] [PubMed]
  92. Wiseman, L.R.; McTavish, D. Formestane. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in the management of breast cancer and prostatic cancer. Drugs 1993, 45, 66–84. [Google Scholar] [CrossRef]
  93. Keating, G.M. Triptorelin embonate (6-month formulation). Drugs 2010, 70, 347–353. [Google Scholar] [CrossRef]
  94. Di Lorenzo, G. Estramustine in prostate cancer: New look at an old drug. Lancet Oncol. 2007, 8, 959–961. [Google Scholar] [CrossRef]
  95. Loke, S.; Liu, L.; Wenzel, M.; Scheffler, H.; Iannone, M.; de la Torre, X.; Schlörer, N.; Botrè, F.; Keiler, A.M.; Bureik, M.; et al. New insights into the metabolism of methyltestosterone and metandienone: Detection of novel A-ring reduced metabolites. Molecules 2021, 26, 1354. [Google Scholar] [CrossRef]
  96. Gordan, G.S.; Wessler, S.; Avioli, L.V. Calusterone in the therapy for advanced breast cancer. JAMA 1972, 219, 483–490. [Google Scholar] [CrossRef] [PubMed]
  97. Ligibel, J.A.; Winer, E.P. Clinical differences among the aromatase inhibitors. Clin. Cancer Res. 2003, 9, 473s–479s. [Google Scholar] [PubMed]
  98. Waddell, W.R.; Kirsch, W.M. Testolactone, sulindac, warfarin, and vitamin K1 for unresectable desmoid tumors. Am. J. Surg. 1991, 161, 416–421. [Google Scholar] [CrossRef]
  99. Gaertner, R.A.; Lewison, E.F.; Finney, G.G., Jr.; Montague, A.C. Combined hormone therapy in advanced breast cancer. Triple-blind study of 68 patients treated with dromostanolone and fluorometholone. Johns Hopkins Med. J. 1968, 123, 138–141. [Google Scholar] [PubMed]
  100. Socas, L.; Zumbado, M.; Pérez-Luzardo, O.; Ramos, A.; Pérez, C.; Hernández, J.R.; Boada, L.D. Hepatocellular adenomas associated with anabolic androgenic steroid abuse in bodybuilders: A report of two cases and a review of the literature. Br. J. Sports Med. 2005, 39, e27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Martin, J.D.; Panagi, M.; Wang, C.; Khan, T.T.; Martin, M.R.; Voutouri, C.; Toh, K.; Papageorgis, P.; Mpekris, F.; Polydorou, C.; et al. Dexamethasone increases cisplatin-loaded nanocarrier delivery and efficacy in metastatic breast cancer by normalizing the tumor microenvironment. ACS Nano 2019, 13, 6396–6408. [Google Scholar] [CrossRef] [PubMed]
  102. Madeddu, C.; Macciò, A.; Panzone, F.; Tanca, F.M.; Mantovani, G. Medroxyprogesterone acetate in the management of cancer cachexia. Expert Opin. Pharmacother. 2009, 10, 1359–1366. [Google Scholar] [CrossRef]
  103. Kim, J.G.; Bae, S.O.; Seo, K.S. A comparison of the effectiveness of complex decongestive physiotherapy and stellate ganglion block with triamcinolone administration in breast cancer-related lymphedema patients. Support Care Cancer 2015, 23, 2305–2310. [Google Scholar] [CrossRef]
  104. Vagos Mata, A.; Espada, E.; Alves, D.; Polo, B.; Costa, M.J.; Lopes, C.; Lacerda, J.F.; Raposo, J. Chronic lymphocytic leukaemia/small lymphocytic lymphoma treatment with rituximab and high-dose methylprednisolone, revisited. Cancer Med. 2021, 10, 8768–8776. [Google Scholar] [CrossRef]
  105. Lewis, D.J.; Duvic, M. Forodesine in the treatment of cutaneous T-cell lymphoma. Expert Opin. Investig. Drugs 2017, 26, 771–775. [Google Scholar] [CrossRef]
  106. Reinhold, U.; Dirschka, T.; Ostendorf, R.; Aschoff, R.; Berking, C.; Philipp-Dormston, W.G.; Hahn, S.; Lau, K.; Jager, A.; Schmitz, B.; et al. A randomized, double-blind, phase III, multicentre study to evaluate the safety and efficacy of BF-200 ALA (Ameluz(R)) vs. placebo in the field-directed treatment of mild-to-moderate actinic keratosis with photodynamic therapy (PDT) when using the BF-RhodoLED(R) lamp. Br. J. Dermatol. 2016, 175, 696–705. [Google Scholar] [PubMed]
  107. Kortuem, K.M.; Stewart, A.K. Carfilzomib. Blood 2013, 121, 893–897. [Google Scholar] [CrossRef] [PubMed]
  108. Ando, K.; Mori, K.; Corradini, N.; Redini, F.; Heymann, D. Mifamurtide for the treatment of nonmetastatic osteosarcoma. Expert Opin. Pharmacother. 2011, 12, 285–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Wood, G.S.; Wu, J. Methotrexate and pralatrexate. Dermatol. Clin. 2015, 33, 747–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Ichikawa, M.; Akimoto, J.; Miki, Y.; Maeda, J.; Takahashi, T.; Fujiwara, Y.; Kohno, M. Photodynamic therapy with talaporfin sodium induces dose- and time-dependent apoptotic cell death in malignant meningioma HKBMM cells. Photodiagnosis Photodyn. Ther. 2019, 25, 29–34. [Google Scholar] [CrossRef] [PubMed]
  111. Vallecorsa, P.; Di Venosa, G.; Gola, G.; Sáenz, D.; Mamone, L.; MacRobert, A.J.; Ramírez, J.; Casas, A. Photodynamic therapy of cutaneous T-cell lymphoma cell lines mediated by 5-aminolevulinic acid and derivatives. J. Photochem. Photobiol. B 2021, 221, 112244. [Google Scholar] [CrossRef]
  112. Mahmoudi, K.; Garvey, K.L.; Bouras, A.; Cramer, G.; Stepp, H.; Jesu Raj, J.G.; Bozec, D.; Busch, T.M.; Hadjipanayis, C.G. 5-aminolevulinic acid photodynamic therapy for the treatment of high-grade gliomas. J. Neurooncol. 2019, 141, 595–607. [Google Scholar] [CrossRef]
  113. Cheng, C.; Michaels, J.; Scheinfeld, N. Alitretinoin: A comprehensive review. Expert Opin. Investig. Drugs 2008, 17, 437–443. [Google Scholar] [CrossRef]
  114. Goodman, G.R.; Beutler, E.; Saven, A. Cladribine in the treatment of hairy-cell leukaemia. Best Pract. Res. Clin. Haematol. 2003, 16, 101–116. [Google Scholar] [CrossRef]
  115. Horikoshi, A.; Takei, K.; Hosokawa, Y.; Sawada, S. The value of oral cytarabine ocfosfate and etoposide in the treatment of refractory and elderly AML patients. Int. J. Hematol. 2008, 87, 118–125. [Google Scholar] [CrossRef]
  116. Sauter, C.; Lamanna, N.; Weiss, M.A. Pentostatin in chronic lymphocytic leukemia. Expert Opin. Drug Metab. Toxicol. 2008, 4, 1217–1222. [Google Scholar] [CrossRef] [PubMed]
  117. Kelemen, E.; Tura, S.; Burger, T. Decreasing risk of leukaemia during prolonged follow-up after mitobronitol therapy for polycythaemia vera. Lancet 1987, 2, 625. [Google Scholar] [CrossRef]
  118. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  119. Liehr, J.G.; Harris, N.J.; Mendoza, J.; Ahmed, A.E.; Giovanella, B.C. Pharmacology of camptothecin esters. Ann. N. Y. Acad. Sci. 2000, 922, 216–223. [Google Scholar] [CrossRef] [PubMed]
  120. Zhao, W.; Zhou, C.; Guan, Z.Y.; Yin, P.; Chen, F.; Tang, Y.J. Structural insights into the inhibition of tubulin by the antitumor agent 4β-(1,2,4-triazol-3-ylthio)-4-deoxypodophyllotoxin. ACS Chem. Biol. 2017, 12, 746–752. [Google Scholar] [CrossRef]
  121. Bonofiglio, D.; Giordano, C.; De Amicis, F.; Lanzino, M.; Andò, S. Natural products as promising antitumoral agents in breast cancer: Mechanisms of action and molecular targets. Min. Rev. Med. Chem. 2016, 18, 596–604. [Google Scholar] [CrossRef]
  122. Tymon-Rosario, J.; Adjei, N.N.; Roque, D.M.; Santin, A.D. Microtubule-interfering drugs: Current and future roles in epithelial ovarian cancer treatment. Cancers 2021, 13, 6239. [Google Scholar] [CrossRef]
  123. Zhang, D.; Kanakkanthara, A. Beyond the paclitaxel and vincaalkaloids: Next generation of plant-derived microtubule-targeting agents with potential anticancer activity. Cancers 2020, 12, 1721. [Google Scholar] [CrossRef]
  124. Khaiwa, N.; Maarouf, N.R.; Darwish, M.H.; Alhamad, D.W.M.; Sebastian, A.; Hamad, M.; Omar, H.A.; Orive, G.; Al-Tel, T.H. Camptothecin’s journey from discovery to WHO essential medicine: Fifty years of promise. Eur. J. Med. Chem. 2021, 223, 113639. [Google Scholar] [CrossRef]
  125. He, W.; Lai, R.; Lin, Q.; Huang, Y.; Wang, L. Arglabin is a plant sesquiterpene lactone that exerts potent anticancer effects on human oral squamous cancer cells via mitochondrial apoptosis and downregulation of the mTOR/PI3K/Akt signaling pathway to inhibit tumor growth in vivo. JBUON 2018, 23, 1679–1685. [Google Scholar]
  126. Burger, T.; Mokoka, T.; Fouche, G.; Steenkamp, P.; Steenkamp, V.; Cordier, W. Solamargine, a bioactive steroidal alkaloid isolated from Solanum aculeastrum induces non-selective cytotoxicity and P-glycoprotein inhibition. BMC Complement. Altern. Med. 2018, 18, 137. [Google Scholar] [CrossRef] [PubMed]
  127. Risdian, C.; Mozef, T.; Wink, J. Biosynthesis of polyketides in Streptomyces. Microorganisms 2019, 7, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Hulst, M.B.; Grocholski, T.; Neefjes, J.J.C.; van Wezel, G.P.; Metsa-Ketela, M. Anthracyclines: Biosynthesis, engineering and clinical applications. Nat. Prod. Rep. 2021, 39, 814–841. [Google Scholar] [CrossRef] [PubMed]
  129. Frederick, C.A.; Williams, L.D.; Ughetto, G.; van der Marel, G.A.; van Boom, J.H.; Rich, A.; Wang, A.H. Structural comparison of anticancer drug-DNA complexes: Adriamycin and daunomycin. Biochemistry 1990, 29, 2538–2549. [Google Scholar] [CrossRef] [PubMed]
  130. Antoniak, S.; Phungphong, S.; Cheng, Z.; Jensen, B.C. Novel mechanisms of anthracycline-induced cardiovascular toxicity: A focus on thrombosis, cardiac atrophy, and programmed cell death. Front. Cardiovasc. Med. 2021, 8, 817977. [Google Scholar] [CrossRef]
  131. Qiao, X.; van der Zanden, S.Y.; Wander, D.P.A.; Borras, D.M.; Song, J.Y.; Li, X.; van Duikeren, S.; van Gils, N.; Rutten, A.; van Herwaarden, T.; et al. Uncoupling DNA damage from chromatin damage to detoxify doxorubicin. Proc. Natl. Acad. Sci. USA 2020, 117, 15182–15192. [Google Scholar] [CrossRef]
  132. Bolzan, A.D.; Bianchi, M.S. DNA and chromosome damage induced by bleomycin in mammalian cells: An update. Mutat. Res. Rev. Mutat. Res. 2018, 775, 51–62. [Google Scholar] [CrossRef]
  133. Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2021 update. Pharmacol. Res. 2021, 165, 105463. [Google Scholar] [CrossRef]
  134. Tan, S.; Li, D.; Zhu, X. Cancer immunotherapy: Pros, cons and beyond. Biomed. Pharmacother. 2020, 124, 109821. [Google Scholar] [CrossRef]
  135. Dhillon, S. Trastuzumab emtansine: A review of its use in patients with HER2-positive advanced breast cancer previously treated with trastuzumab-based therapy. Drugs 2014, 74, 675–686. [Google Scholar] [CrossRef]
  136. Buckel, L.; Savariar, E.N.; Crisp, J.L.; Jones, K.A.; Hicks, A.M.; Scanderbeg, D.J.; Nguyen, Q.T.; Sicklick, J.K.; Lowy, A.M.; Tsien, R.Y.; et al. Tumor radiosensitization by monomethyl auristatin E: Mechanism of action and targeted delivery. Cancer Res. 2015, 75, 1376–1387. [Google Scholar] [CrossRef] [PubMed]
  137. Kantarjian, H.; Ravandi, F.; Short, N.J.; Huang, X.; Jain, N.; Sasaki, K.; Daver, N.; Pemmaraju, N.; Khoury, J.D.; Jorgensen, J.; et al. Inotuzumab ozogamicin in combination with low-intensity chemotherapy for older patients with Philadelphia chromosome-negative acute lymphoblastic leukaemia: A single-arm, phase 2 study. Lancet Oncol. 2018, 19, 240–248. [Google Scholar] [CrossRef]
  138. Thara, E.; Gitlitz, B.J. Eribulin: A new-generation antimicrotubule agent in lung cancer therapy. Future Oncol. 2014, 10, 1913–1924. [Google Scholar] [CrossRef] [PubMed]
  139. Dang, J.; Fu, J.; Zhang, Z.; Liu, D.; Cheng, D.; Fan, H. Comparison between trabectedin and doxorubicin in soft-tissue sarcomas: A systematic review and meta-analysis. Ann. Transl. Med. 2021, 9, 1764. [Google Scholar] [CrossRef]
  140. Losada, A.; Berlanga, J.J.; Molina-Guijarro, J.M.; Jimenez-Ruiz, A.; Gago, F.; Aviles, P.; de Haro, C.; Martinez-Leal, J.F. Generation of endoplasmic reticulum stress and inhibition of autophagy by plitidepsin induces proteotoxic apoptosis in cancer cells. Biochem. Pharmacol. 2020, 172, 113744. [Google Scholar] [CrossRef]
  141. Patel, S.; Petty, W.J.; Sands, J.M. An overview of lurbinectedin as a new second-line treatment option for small cell lung cancer. Ther. Adv. Med. Oncol. 2021, 13, 17588359211020529. [Google Scholar] [CrossRef]
  142. Jiménez, C. Marine natural products in medicinal chemistry. Front. Oncol. 2018, 9, 959–961. [Google Scholar] [CrossRef] [Green Version]
  143. Blunt, J.W.; Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2018, 35, 8–53. [Google Scholar] [CrossRef] [Green Version]
  144. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2021, 38, 362–413. [Google Scholar] [CrossRef]
  145. Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2022, 39, 1122–1171. [Google Scholar] [CrossRef]
  146. Keiler, A.M.; Zierau, O.; Wolf, S.; Diel, P.; Schänzer, W.; Vollmer, G.; Machalz, D.; Wolber, G.; Parr, M.K. Androgen- and estrogen-receptor mediated activities of 4-hydroxytestosterone, 4-hydroxyandrostenedione and their human metabolites in yeast based assays. Toxicol. Lett. 2018, 292, 39–45. [Google Scholar] [CrossRef] [PubMed]
  147. Sardela, V.F.; Sardela, P.D.O.; Lisboa, R.R.; Matias, B.F.; Anselmo, C.S.; de Carvalho, A.R.; Nunes, I.K.C. Comprehensive zebrafish water tank experiment for metabolic studies of testolactone. Zebrafish 2020, 17, 104–111. [Google Scholar] [CrossRef] [PubMed]
  148. Suttmann, H.; Gleissner, J.; Huebner, A.; Mathes, T.; Baurecht, W.; Krützfeldt, K.; Sweiti, H.; Feyerabend, S. Adherence measures for patients with metastatic castration-resistant prostate cancer treated with abiraterone acetate plus prednisone: Results of a prospective, cluster-randomized trial. Cancers 2020, 12, 2550. [Google Scholar] [CrossRef] [PubMed]
  149. Rammohan, K.; Coyle, P.K.; Sylvester, E.; Galazka, A.; Dangond, F.; Grosso, M.; Leist, T.P. The development of cladribine tablets for the treatment of multiple sclerosis: A comprehensive review. Drugs 2020, 80, 1901–1928. [Google Scholar] [CrossRef]
  150. Brosa, M.; García del Muro, X.; Mora, J.; Villacampa, A.; Pozo-Rubio, T.; Cubells, L.; Montoto, C. Orphan drugs revisited cost-effectiveness analysis of the addition of mifamurtide to the conventional treatment of osteosarcoma. Expert Rev. Pharmacoecon. Outcomes Res. 2015, 15, 331–340. [Google Scholar] [CrossRef]
  151. Yee, A.J. The role of carfilzomib in relapsed/refractory multiple myeloma. Ther. Adv. Hematol. 2021, 12, 1–13. [Google Scholar] [CrossRef]
  152. Hoy, S.M. Carfilzomib triple combination therapy: A review in relapsed multiple myeloma. Target Oncol. 2016, 11, 255–262. [Google Scholar] [CrossRef]
  153. Sjoberg, H.T.; Philippou, Y.; Magnussen, A.L.; Tullis, I.D.C.; Bridges, E.; Chatrian, A.; Lefebvre, J.; Tam, K.H.; Murphy, E.A.; Rittscher, J.; et al. Tumour irradiation combined with vascular-targeted photodynamic therapy enhances antitumour effects in pre-clinical prostate cancer. Br. J. Cancer 2021, 125, 534–546. [Google Scholar] [CrossRef]
  154. Regillo, C.D. Update on photodynamic therapy. Curr. Opin. Ophthalmol. 2000, 11, 166–170. [Google Scholar] [CrossRef]
  155. He, J.; Liu, C.; Li, T.; Liu, Y.; Wang, S.; Zhang, J.; Chen, L.; Wang, C.; Feng, Y.; Floris, G.; et al. Pictorial imaging-histopathology correlation in a rabbit with hepatic VX2 tumor treated by transarterial vascular disrupting agent administration. Int. J. Med. Sci. 2020, 17, 2269–2275. [Google Scholar] [CrossRef]
  156. Suzuki, T.; Tanaka, M.; Sasaki, M.; Ichikawa, H.; Nishie, H.; Kataoka, H. Vascular shutdown by photodynamic therapy using talaporfin sodium. Cancers 2020, 12, 2369. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, X.; Gao, C.; Guo, L.; Hu, G.; Luo, Q.; Liu, J.; Nielsen, J.; Chen, J.; Liu, L. DCEO biotechnology: Tools to design, construct, evaluate, and optimize the metabolic pathway for biosynthesis of chemicals. Chem. Rev. 2018, 118, 4–72. [Google Scholar] [CrossRef]
  158. Weber, T.; Blin, K.; Duddela, S.; Krug, D.; Kim, H.U.; Bruccoleri, R.; Lee, S.Y.; Fischbach, M.A.; Muller, R.; Wohlleben, W.; et al. antiSMASH 3.0-a comprehensive resource for the genome mining of biosynthetic gene clusters. Nucleic Acids Res. 2015, 43, W237–W243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Blin, K.; Wolf, T.; Chevrette, M.G.; Lu, X.; Schwalen, C.J.; Kautsar, S.A.; Suarez Duran, H.G.; de Los Santos, E.L.C.; Kim, H.U.; Nave, M.; et al. antiSMASH 4.0-improvements in chemistry prediction and gene cluster boundary identification. Nucleic Acids Res. 2017, 45, W36–W41. [Google Scholar] [CrossRef] [PubMed]
  160. Medema, M.H.; Blin, K.; Cimermancic, P.; de Jager, V.; Zakrzewski, P.; Fischbach, M.A.; Weber, T.; Takano, E.; Breitling, R. antiSMASH: Rapid identification, annotation and analysis of secondary metabolite biosynthesis gene clusters in bacterial and fungal genome sequences. Nucleic Acids Res. 2011, 39, W339–W346. [Google Scholar] [CrossRef] [PubMed]
  161. Blin, K.; Kim, H.U.; Medema, M.H.; Weber, T. Recent development of antiSMASH and other computational approaches to mine secondary metabolite biosynthetic gene clusters. Brief Bioinform. 2019, 20, 1103–1113. [Google Scholar] [CrossRef]
  162. Machado, H.; Tuttle, R.N.; Jensen, P.R. Omics-based natural product discovery and the lexicon of genome mining. Curr. Opin. Microbiol. 2017, 39, 136–142. [Google Scholar] [CrossRef] [Green Version]
  163. Zhang, D.; Wang, X.X.; Ye, Y.; He, Y.; He, F.Q.; Tian, Y.Q.; Luo, Y.Z.; Liang, S.F. Label-free proteomic dissection on dptP-deletion mutant uncovers dptP involvement in strain growth and daptomycin tolerance of Streptomyces roseosporus. Microb. Biotechnol. 2021, 14, 708–725. [Google Scholar] [CrossRef]
  164. Norsigian, C.J.; Pusarla, N.; McConn, J.L.; Yurkovich, J.T.; Drager, A.; Palsson, B.O.; King, Z. BiGG Models 2020: Multi-strain genome-scale models and expansion across the phylogenetic tree. Nucleic Acids Res. 2020, 48, D402–D406. [Google Scholar] [CrossRef]
  165. Sulheim, S.; Kumelj, T.; van Dissel, D.; Salehzadeh-Yazdi, A.; Du, C.; van Wezel, G.P.; Nieselt, K.; Almaas, E.; Wentzel, A.; Kerkhoven, E.J. Enzyme-constrained models and omics analysis of Streptomyces coelicolor reveal metabolic changes that enhance heterologous production. iScience 2020, 23, 101525. [Google Scholar] [CrossRef]
  166. Cotner, M.; Zhan, J.; Zhang, Z. A computational metabolic model for engineered production of resveratrol in Escherichia coli. ACS Synth. Biol. 2021, 10, 1992–2001. [Google Scholar] [CrossRef] [PubMed]
  167. Cardoso, J.G.R.; Jensen, K.; Lieven, C.; Laerke Hansen, A.S.; Galkina, S.; Beber, M.; Ozdemir, E.; Herrgard, M.J.; Redestig, H.; Sonnenschein, N. Cameo: A python library for computer aided metabolic engineering and optimization of cell factories. ACS Synth. Biol. 2018, 7, 1163–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Luo, Y.; Enghiad, B.; Zhao, H. New tools for reconstruction and heterologous expression of natural product biosynthetic gene clusters. Nat. Prod. Rep. 2016, 33, 174–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Gibson, D.G.; Young, L.; Chuang, R.Y.; Venter, J.C.; Hutchison, C.A., 3rd; Smith, H.O. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 2009, 6, 343–345. [Google Scholar] [CrossRef] [PubMed]
  170. Liu, S.H.; Wang, W.; Wang, K.B.; Zhang, B.; Li, W.; Shi, J.; Jiao, R.H.; Tan, R.X.; Ge, H.M. Heterologous expression of a cryptic giant type I PKS gene cluster leads to the production of ansaseomycin. Org. Lett. 2019, 21, 3785–3788. [Google Scholar] [CrossRef]
  171. Jiang, W.; Zhao, X.; Gabrieli, T.; Lou, C.; Ebenstein, Y.; Zhu, T.F. Cas9-Assisted targeting of chromosome segments CATCH enables one-step targeted cloning of large gene clusters. Nat. Commun. 2015, 6, 8101. [Google Scholar] [CrossRef] [Green Version]
  172. Wang, H.; Li, Z.; Jia, R.; Hou, Y.; Yin, J.; Bian, X.; Li, A.; Muller, R.; Stewart, A.F.; Fu, J.; et al. RecET direct cloning and Redalphabeta recombineering of biosynthetic gene clusters, large operons or single genes for heterologous expression. Nat. Protoc. 2016, 11, 1175–1190. [Google Scholar] [CrossRef]
  173. Chen, H.; Li, M.; Liu, C.; Zhang, H.; Xian, M.; Liu, H. Correction to: Enhancement of the catalytic activity of isopentenyl diphosphate isomerase (IDI) from Saccharomyces cerevisiae through random and site-directed mutagenesis. Microb. Cell Fact. 2020, 19, 8. [Google Scholar] [CrossRef]
  174. Tong, Y.; Whitford, C.M.; Robertsen, H.L.; Blin, K.; Jørgensen, T.S.; Klitgaard, A.K.; Gren, T.; Jiang, X.; Weber, T.; Lee, S.Y. Highly efficient DSB-free base editing for streptomycetes with CRISPR-BEST. Proc. Natl. Acad. Sci. USA. 2019, 116, 20366–20375. [Google Scholar] [CrossRef] [Green Version]
  175. Kim, Y.B.; Komor, A.C.; Levy, J.M.; Packer, M.S.; Zhao, K.T.; Liu, D.R. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 2017, 35, 371–376. [Google Scholar] [CrossRef]
  176. Eid, A.; Alshareef, S.; Mahfouz, M.M. CRISPR base editors: Genome editing without double-stranded breaks. Biochem. J. 2018, 475, 1955–1964. [Google Scholar] [CrossRef]
  177. Xia, H.; Li, X.; Li, Z.; Zhan, X.; Mao, X.; Li, Y. The application of regulatory cascades in Streptomyces: Yield ehancement and metabolite mining. Front. Microbiol. 2020, 11, 406. [Google Scholar] [CrossRef] [Green Version]
  178. Wang, Y.; Cobb, R.E.; Zhao, H. High-efficiency genome editing of Streptomyces species by an engineered CRISPR/Cas system. Methods Enzymol. 2016, 575, 271–284. [Google Scholar]
  179. Liu, H.; Tian, Y.; Zhou, Y.; Kan, Y.; Wu, T.; Xiao, W.; Luo, Y. Multi-modular engineering of Saccharomyces cerevisiae for high-titre production of tyrosol and salidroside. Microb. Biotechnol. 2021, 14, 2605–2616. [Google Scholar] [CrossRef]
  180. Tan, G.Y.; Deng, K.; Liu, X.; Tao, H.; Chang, Y.; Chen, J.; Chen, K.; Sheng, Z.; Deng, Z.; Liu, T. Heterologous biosynthesis of spinosad: An omics-guided large polyketide synthase gene cluster reconstitution in Streptomyces. ACS Synth. Biol. 2017, 6, 995–1005. [Google Scholar] [CrossRef] [PubMed]
  181. Xu, J.; Zhang, J.; Zhuo, J.; Li, Y.; Tian, Y.; Tan, H. Activation and mechanism of a cryptic oviedomycin gene cluster via the disruption of a global regulatory gene, adpA, in Streptomyces ansochromogenes. J. Biol. Chem. 2017, 292, 19708–19720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Rutledge, P.J.; Challis, G.L. Discovery of microbial natural products by activation of silent biosynthetic gene clusters. Nat. Rev. Microbiol. 2015, 13, 509–523. [Google Scholar] [CrossRef]
  183. Kudo, K.; Hashimoto, T.; Hashimoto, J.; Kozone, I.; Kagaya, N.; Ueoka, R.; Nishimura, T.; Komatsu, M.; Suenaga, H.; Ikeda, H.; et al. In vitro Cas9-assisted editing of modular polyketide synthase genes to produce desired natural product derivatives. Nat. Commun. 2020, 11, 4022. [Google Scholar] [CrossRef] [PubMed]
  184. Myronovskyi, M.; Rosenkranzer, B.; Nadmid, S.; Pujic, P.; Normand, P.; Luzhetskyy, A. Generation of a cluster-free Streptomyces albus chassis strains for improved heterologous expression of secondary metabolite clusters. Metab. Eng. 2018, 49, 316–324. [Google Scholar] [CrossRef] [PubMed]
  185. Bu, Q.T.; Yu, P.; Wang, J.; Li, Z.Y.; Chen, X.A.; Mao, X.M.; Li, Y.Q. Rational construction of genome-reduced and high-efficient industrial Streptomyces chassis based on multiple comparative genomic approaches. Microb. Cell Fact. 2019, 18, 16. [Google Scholar] [CrossRef]
  186. Lyu, H.N.; Liu, H.W.; Keller, N.P.; Yin, W.B. Harnessing diverse transcriptional regulators for natural product discovery in fungi. Nat. Prod. Rep. 2019, 37, 6–16. [Google Scholar] [CrossRef] [PubMed]
  187. Jin, L.Q.; Jin, W.R.; Ma, Z.C.; Shen, Q.; Cai, X.; Liu, Z.Q.; Zheng, Y.G. Promoter engineering strategies for the overproduction of valuable metabolites in microbes. Appl. Microbiol. Biotechnol. 2019, 103, 8725–8736. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, B.; Guo, F.; Dong, S.H.; Zhao, H. Activation of silent biosynthetic gene clusters using transcription factor decoys. Nat. Chem. Biol. 2019, 15, 111–114. [Google Scholar] [CrossRef] [PubMed]
  189. Ho, H.I.; Fang, J.R.; Cheung, J.; Wang, H.H. Programmable CRISPR-Cas transcriptional activation in bacteria. Mol. Syst. Biol. 2020, 16, e9427. [Google Scholar] [CrossRef] [PubMed]
  190. Vickers, C.E.; Blank, L.M.; Kromer, J.O. Grand challenge commentary: Chassis cells for industrial biochemical production. Nat. Chem. Biol. 2010, 6, 875–877. [Google Scholar] [CrossRef] [PubMed]
  191. Zhu, F.; Qin, C.; Tao, L.; Liu, X.; Shi, Z.; Ma, X.; Jia, J.; Tan, Y.; Cui, C.; Lin, J.; et al. Clustered patterns of species origins of nature-derived drugs and clues for future bioprospecting. Proc. Natl. Acad. Sci. USA 2011, 108, 12943–12948. [Google Scholar] [CrossRef] [Green Version]
  192. Engels, B.; Dahm, P.; Jennewein, S. Metabolic engineering of taxadiene biosynthesis in yeast as a first step towards Taxol (Paclitaxel) production. Metab. Eng. 2008, 10, 201–206. [Google Scholar] [CrossRef]
  193. Jiang, M.; Stephanopoulos, G.; Pfeifer, B.A. Toward biosynthetic design and implementation of Escherichia coli-derived paclitaxel and other heterologous polyisoprene compounds. Appl. Environ. Microbiol. 2012, 78, 2497–2504. [Google Scholar] [CrossRef] [Green Version]
  194. Ajikumar, P.K.; Xiao, W.H.; Tyo, K.E.; Wang, Y.; Simeon, F.; Leonard, E.; Mucha, O.; Phon, T.H.; Pfeifer, B.; Stephanopoulos, G. Isoprenoid pathway optimization for taxol precursor overproduction in Escherichia coli. Science 2010, 330, 70–74. [Google Scholar] [CrossRef] [Green Version]
  195. Maeda, H.A. Harnessing evolutionary diversification of primary metabolism for plant synthetic biology. J. Biol. Chem. 2019, 294, 16549–16566. [Google Scholar] [CrossRef] [Green Version]
  196. Zhang, J.; Chen, Y.; Fu, L.; Guo, E.; Wang, B.; Dai, L.; Si, T. Accelerating strain engineering in biofuel research via build and test automation of synthetic biology. Curr. Opin. Biotechnol. 2021, 67, 88–98. [Google Scholar] [CrossRef] [PubMed]
  197. Chao, R.; Mishra, S.; Si, T.; Zhao, H. Engineering biological systems using automated biofoundries. Metab. Eng. 2017, 42, 98–108. [Google Scholar] [CrossRef] [PubMed]
  198. HamediRad, M.; Chao, R.; Weisberg, S.; Lian, J.; Sinha, S.; Zhao, H. Towards a fully automated algorithm driven platform for biosystems design. Nat. Commun. 2019, 10, 5150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Parakh, S.; Parslow, A.C.; Gan, H.K.; Scott, A.M. Antibody-mediated delivery of therapeutics for cancer therapy. Expert Opin. Drug Deliv. 2016, 13, 401–419. [Google Scholar] [CrossRef]
  200. Huang, L.; Zhao, S.; Fang, F.; Xu, T.; Lan, M.; Zhang, J. Advances and perspectives in carrier-free nanodrugs for cancer chemo-monotherapy and combination therapy. Biomaterials 2021, 268, 120557. [Google Scholar] [CrossRef] [PubMed]
  201. Deeks, E.D. Polatuzumab vedotin: First global approval. Drugs 2019, 79, 1467–1475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Diefenbach, C.; Kahl, B.S.; McMillan, A.; Briones, J.; Banerjee, L.; Cordoba, R.; Miall, F.; Burke, J.M.; Hirata, J.; Jiang, Y.; et al. Polatuzumab vedotin plus obinutuzumab and lenalidomide in patients with relapsed or refractory follicular lymphoma: A cohort of a multicentre, single-arm, phase 1b/2 study. Lancet Haematol. 2021, 8, e891–e901. [Google Scholar] [CrossRef]
  203. He, Y.; Luo, Y.; Huang, L.; Zhang, D.; Wang, X.; Ji, J.; Liang, S. New frontiers against sorafenib resistance in renal cell carcinoma: From molecular mechanisms to predictive biomarkers. Pharmacol. Res. 2021, 170, 105732. [Google Scholar] [CrossRef]
Figure 1. All approved anticancer drugs derived from natural products from 1950 to 2019. (A) The sources of anticancer drugs approved from 1950_2019 and their proportions. N: natural product. ND: natural product derivative. B: biological macromolecule. S: synthetic drug. NM: mimic of natural product. S*: synthetic drug with NP pharmacophore. V: vaccine. (B,C) Anticancer drugs derived from natural products. (D) All approved anticancer natural products by source/year. ADC: antibody drug conjugate.
Figure 1. All approved anticancer drugs derived from natural products from 1950 to 2019. (A) The sources of anticancer drugs approved from 1950_2019 and their proportions. N: natural product. ND: natural product derivative. B: biological macromolecule. S: synthetic drug. NM: mimic of natural product. S*: synthetic drug with NP pharmacophore. V: vaccine. (B,C) Anticancer drugs derived from natural products. (D) All approved anticancer natural products by source/year. ADC: antibody drug conjugate.
Molecules 27 07480 g001
Figure 2. Chemical structures for plant-derived (A), ADCs (B), marine (C), hormones and other natural products (D) and microorganism-derived natural products (E).
Figure 2. Chemical structures for plant-derived (A), ADCs (B), marine (C), hormones and other natural products (D) and microorganism-derived natural products (E).
Molecules 27 07480 g002
Figure 4. Bioinformatic analyses for natural product improvement. (A) Extracting the target information via online databases. (B) Discovering new natural products with the help of omics analyses. (C) Computer modeling predictions via metabolic networks.
Figure 4. Bioinformatic analyses for natural product improvement. (A) Extracting the target information via online databases. (B) Discovering new natural products with the help of omics analyses. (C) Computer modeling predictions via metabolic networks.
Molecules 27 07480 g004
Figure 5. Tools for pathway reconstruction or engineering of natural product. (A) DNA assembly tools for assembling multiple DAN fragments of biosynthetic gene cluster in vivo or in vitro. (B) Capturing biosynthetic gene clusters of natural products by direct cloning. (C) Genome editing for improving natural products through single-base editing, gene deletion, gene insertion and module editing using CRISPR tools.
Figure 5. Tools for pathway reconstruction or engineering of natural product. (A) DNA assembly tools for assembling multiple DAN fragments of biosynthetic gene cluster in vivo or in vitro. (B) Capturing biosynthetic gene clusters of natural products by direct cloning. (C) Genome editing for improving natural products through single-base editing, gene deletion, gene insertion and module editing using CRISPR tools.
Molecules 27 07480 g005
Figure 6. Biosystems for developing natural products. (A) Cell factory construction via genome mining, transcription regulation, metabolic engineering, protein engineering and transporter engineering. (B) Application of artificial intelligence in natural product improvement. Researcher writes programs and builds workstations, and robot accomplishes the instructions. In the last, researcher analyzes the data.
Figure 6. Biosystems for developing natural products. (A) Cell factory construction via genome mining, transcription regulation, metabolic engineering, protein engineering and transporter engineering. (B) Application of artificial intelligence in natural product improvement. Researcher writes programs and builds workstations, and robot accomplishes the instructions. In the last, researcher analyzes the data.
Molecules 27 07480 g006
Table 1. All natural anticancer drugs approved from 1950 to 2021.
Table 1. All natural anticancer drugs approved from 1950 to 2021.
Active IngredientDrug NameTarget Cancer TypeYear Intro.SourceRef.
plant NPs
asparaginase
Erwinia chrysanthemi
Rylazeacute lymphoblastic leukemia and lymphoblastic lymphoma2021Erwinia chrysanthemi[31]
padeliporfin
potassium
Tookadprostate cancer2015a derivative of chlorophyll[32]
paclitaxel injection
concentrate for
nanodispersion
PICNmetastatic breast cancer2014taxus species[33]
homoharringtonine Ceflatonin myeloid leukemia2012Cephalotaxus harringtonia[34]
ingenol mebutate Picato actinic keratosis2012Euphorbia peplu[35]
cabazitaxel Jevtana castration-resistant metastatic prostate cancer2010a taxane derivative [36]
vinflunine Javlor advanced and metastatic urothelial carcinoma2010a semisynthetic vinca-alkaloid[37]
nanoparticle-based formulation of paclitaxel Nanoxel ovarian, non-small cell lung, breast, gastric, endometrial and pancreatic cancers2007taxus species[38]
albumin-bound paclitaxelAbraxane ovarian, non-small cell lung, breast, gastric, endometrial and pancreatic cancers2005taxus species[39,40]
belotecan HCL Camtobell small cell lung cancer 2004a camptothecin analog[41]
liposomal
formulation of paclitaxel
Lipusu non-small cell lung, breast, gastric,
endometrial and pancreatic cancers
2003taxus species[42]
fulvestrant Faslodex advanced breast cancer2002a taxane plant product[43]
arglabin n.r.oral squamous cell carcinoma, breast cancer1999Artemisia species[44]
topotecan HCl Hycamptin small cell lung cancer1996a camptothecin analog [45]
etoposide
phosphate
Etopophos advanced-stage Hodgkin lymphoma1996a semisynthetic derivative of podophyllotoxin [46]
docetaxel Taxotere HER2-positive metastatic breast cancer 1995yew tree[47]
irinotecan HCl Campto colorectal and pancreatic cancer1994a camptothecin analog[48]
paclitaxel Taxol non-small cell lung, breast, gastric, endometrial and pancreatic cancers1993pacific yew trees[49]
masoprocol Actinex a potent sensitizer1992creosote bush[50]
vinorelbine Navelbine non-small cell lung cancer1989a semisynthetic vinca alkaloid[51]
solamarginesCuraderm nasopharyngeal carcinoma cells1989Solanum undatum[52]
elliptinium acetate Celiptiumbreast cancer1983a derivative of ellipticine derived from plant[53]
etoposiden.r.extensive-stage small cell lung cancer1980a semisynthetic derivative of podophyllotoxin [54]
vindesinen.r.Leukemia, non-small-cell lung cancer1979Catharanthus roseus[55]
teniposiden.r.acute lymphoblastic leukemia1967a semisynthetic derivative of podophyllotoxin [56]
vinblastinen.r.Hodgkin lymphoma1965Catharanthus roseus[57]
vincristinen.r.hematologic malignancies and solid tumors1963nerium oleander[57]
microbial NPs
midostaurin Rydapt acute myeloid leukemia, advanced systemic mastocytosis2017a derivate of staurosporine produced by Streptomyces staurosporeus[58]
romidepsin Istodax T cell lymphomas2010Chromobacterium violaceum[59]
temsirolimus Toricel relapsed or refractory solid tumors2007a derivative of sirolimus produced by Streptomyces hygroscopicus[60]
ixabepilone Ixempra breast cancer2007a derivate of epothilone B produced by Sorangium cellulosum [61]
amrubicin HCl Calsed small-cell lung cancer 2002an anthracyclin analogue[62]
valrubicin Valstar non-muscle invasive bladder cancer1999a derivative of the anthracycline doxorubicin[63]
zinostatin stimalamer Smancs hepatocellular
carcinoma
1994chemically synthesized by coupling one molecule of neocarzinostatin produced by Streptomyces carzinostaticus[64]
idarubicin HCl Zavedos acute myelogenous leukemia1990an analogue of daunorubicin produced by Streptomyces peucetius[65]
pirarubicinPinorubicin lung cancer, breast cancer1988a novel anthracycline derivative of doxorubicin [66]
epirubicin HCI Farmorubicin B cell lymphoma, head and neck
cancer and other solid cancers
1984a semisynthetic derivative of doxorubicin [67]
aclarubicin Aclacin acute myeloid leukemia, hematologic cancers and solid tumors1981Streptomyces galilacus[68]
peplomycin Pepleo cutaneous squamous cell carcinoma, prostatic cancer, breast cancer1981a derivative of bleomycin[69]
asparaginasen.r.leukemia and lymphoma1969Escherichia coli[70]
daunomycinn.r.acute promyelocytic1967Streptomyces species[71]
bleomycinn.r.squamous cell carcinoma from head and necki, lymphomas, testicular
carcinoma
1966Streptomyces verticillus[72]
doxorubicinn.r.ladder, breast, stomach, lung, ovaries, thyroid, soft tissue sarcoma1966Streptomyces peucetius var. caesius[73]
actinomycin Dn.r.solid tumors in children and
choriocarcinoma in adult women
1964Streptomyces species[74]
mithramycinn.r.chronic and acute myeloid leukemia, testicular carcinoma1961Streptomyces species[75]
mitomycin Cn.r.bladder, breast carcinoma, head and neck malignancies, and some other gastrointestinal cancer1956Streptomyces caespitosus[76]
leucovorinn.r.haematologic malignancies and
osteosarcomas
1950Leuconostoc citrovorum[77]
antibody-conjugated NPs
enfortumab
vedotin-ejfv
Padcevrefractory bladder cancer2019a nectin-4-targeted antibody conjugated to monomethyl auristatin E, which is a toxin[78]
polatuzumab
vedotin
Polivy relapsed or refractory diffuse large
B-cell lymphoma
2019an anti-CD79b antibody conjugated to monomethyl auristatin E, which is a toxin[79]
trastuzumab
emtansine
Kadcyla HER2-positive early breast cancer 2019a monoclonal antibody trastuzumaba conjugated to emtansine, which is a derivative of maytansine[80]
inotuzumab
ozogamicin
Mundesine acute lymphoblastic leukemia2017an anti-22 antibody linked to
calicheamicin
[81]
brentuximab
vedotin
Adcetris relapsed or refractory hodgkin
lymphoma
2011an anti-CD30 antibody conjugated to monomethyl auristatin E, which is a toxin[82]
gemtuzumab
ozogamicin
Mylotarg acute myeloid leukemia 2000anti-CD33 antibody linked to calicheamicin, a toxin[83]
marine NPs
lurbinectedinZepzelcametastatic small cell lung cancer2020a marine-derived drug[84]
aplidine Aplidin acute lymphoblastic leukemia2018Aplidium albican[85]
trabectedin Yondelissoft tissue sarcomas—liposarcoma
and leiomyosarcoma
2015Ecteinascidia turbinata[86]
eribulin Halaven breast cancer and soft-tissue sarcoma2010a macrocyclic ketone analogue of the halichondrin[87]
hormones
abiraterone acetate Zytiga castration-resistant prostate cancer2011a semisynthetic steroid[88]
vapreotide acetate Docrised esophageal variceal bleeding2004a somatostatin analogue[89]
exemestane Aromasin hormone receptor-positive breast
cancer
1999endocrine agent, steroidal compound [90]
angiotensin II Delivert pancreatic cancer1994an endogenous hormone [91]
formestane Lentaron breast cancer, prostatic cancer1993a steroid substrate analog[92]
triptorelin Decapeptyl prostate cancer1986a decapeptide analog of luteinizing hormone releasing hormone[93]
estramustinen.r.prostate cancer1980a stable estradiol [94]
methyltestosteronen.r.breast cancer1974an anabolic–androgenic steroid[95]
calusteronen.r.advanced breast cancer1973an androgenic steroid[96]
megesterol acetaten.r.metastatic breast cancer1971n.r.[97]
testolactonen.r.desmoid tumors, breast cancer1969a nonselective steroid[98]
dromostanolonen.r.breast cancer1961an androgen steroid[99]
nandrolone
phenylpropionate
n.r.hepatocellular adenomas1959a steroid[100]
dexamethasonen.r.breast cancer, acute leukemia
lymphoma
1958a glucocorticoid[101]
medroxyprogesterone acetaten.r.hormone-related cancers, cachexia syndrome1958a hormone progesterone variant[102]
triamcinolonen.r.ocular conditions unresponsive to topical steroids1958a corticosteroid[103]
methylprednisolonen.r.hodgkin lymphoma1955a corticosteroid[104]
other NPs
forodesine HCl Mundesine relapsed peripheral T cell lymphoma2017a purine nucleoside analogue[105]
a nanoemulsion formulation containing 10% aminolaevulinic acid hydrochlorideAmeluz lesion-directed and field-directed actinic keratosis2012the precursor of the endogenous photosensitizer Protoporphyrin IX[106]
carfilzomib Kyprolis relapsed or refractory multiple myeloma2012a peptide epoxyketone [107]
mifamurtide Junovan nonmetastatic osteosarcoma2010a conjugate of muramyl tripeptide linked to dipalmitoyl phosphatidyl ethanolamine[108]
pralatrexate Folotynrelapsed/refractory peripheral T cell lymphomas2009a folic acid analogue[109]
talaporfin sodium Laserphyrinesophageal cancer2004mono-L-asparthyl chlorine e6: NPe-6[110]
methyl
aminolaevulinate
Metvixhigh-risk basal cell carcinoma2001the precursor of the endogenous photosensitizer Protoporphyrin IX[111]
aminolevulinic acid Levulan premalignant and malignant diseases2000the first metabolite in the heme biosynthesis pathway [112]
alitretinoin Panretin acute promyelocytic leukemia1999an endogenous vitamin A derivative, 9-cis-retinoic acid[113]
cladribine Leustatin hairy-cell leukaemia1993a purine nucleoside analogue [114]
cytarabine ocfosfate Starsaid acute myeloid leukemia1993an orally applicable prodrug of cytosine arabinoside[115]
pentostatin Nipent lymphocytic leukemia1992an analogue of purine[116]
mitobronitoln.r.acute myeloblastic leukemia1979n.r.[117]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, H.; He, Y.; Jian, M.; Fu, X.; Cheng, Y.; He, Y.; Fang, J.; Li, L.; Zhang, D. Breaking the Bottleneck in Anticancer Drug Development: Efficient Utilization of Synthetic Biology. Molecules 2022, 27, 7480. https://doi.org/10.3390/molecules27217480

AMA Style

Wang H, He Y, Jian M, Fu X, Cheng Y, He Y, Fang J, Li L, Zhang D. Breaking the Bottleneck in Anticancer Drug Development: Efficient Utilization of Synthetic Biology. Molecules. 2022; 27(21):7480. https://doi.org/10.3390/molecules27217480

Chicago/Turabian Style

Wang, Haibo, Yu He, Meiling Jian, Xingang Fu, Yuheng Cheng, Yujia He, Jun Fang, Lin Li, and Dan Zhang. 2022. "Breaking the Bottleneck in Anticancer Drug Development: Efficient Utilization of Synthetic Biology" Molecules 27, no. 21: 7480. https://doi.org/10.3390/molecules27217480

Article Metrics

Back to TopTop