Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 1.
Published in final edited form as: Curr Protoc Pharmacol. 2019 Sep;86(1):e67. doi: 10.1002/cpph.67

Natural Products as a Foundation for Drug Discovery

John A Beutler 1
PMCID: PMC7442317  NIHMSID: NIHMS1608203  PMID: 31539923

Abstract

Many natural products have been used as drugs for the treatment of diverse indications. While most U.S. pharmaceutical companies have over the years reduced or eliminated their in-house natural product groups, new approaches for compound screening and chemical synthesis are resurrecting interest in exploring the therapeutic value of natural products. The aim of this report is to review emerging strategies and techniques that have made natural products a viable strategic choice for inclusion in drug discovery programs.

Keywords: natural products, drugs discovery, HTS

1. History

1.1. Early natural product drugs

Humans have long used naturally occurring substances for medical purposes. In particular, plant products have played a leading medical role in most cultures. With the development of the science of chemistry in the early 19th century, it became possible to examine plant materials more closely to identify biologically active constituents and to understand their mechanisms of action. In 1804 Sertürner purified morphine from the opium poppy and found that it largely reproduced opium’s analgesic and sedative effects (Lockemann, 1951). His success led others to seek “active principles” of medicinal plants. During the 19th century bioactive natural products were purified from cinchona (quinine) (Pelletier and Caventou, 1820, Borchardt, 1996), coca (cocaine) (Gaedcke, 1855, Gay et al., 1975) and many other plants. The ability to determine chemical structures developed later, with the planar structure of cocaine first reported in 1898 (Willstätter and Müller, 1898) of quinine in 1908 (Rabe, 1908) and morphine in 1923 (Gulland and Robinson, 1923). The technology needed to synthesize these evolved mainly in the 20th century, with morphine being synthesized in 1956 (Gates and Tschudi, 1956). While the active principle hypothesis has not provided an explanation for all biological activities of natural substances, it has been the most fruitful approach for addressing this issue.

1.2. The antibiotic era

The identification by Fleming (Fleming, 1929) of the antibacterial properties of penicillin and its subsequent isolation by Chain and Florey (Chain et al., 1940) revolutionized the treatment of bacterial infections. This led to extensive screening of microbes, particularly soil actinomycetes and fungi, in an attempt to identify other antibiotics. Using simple bioassays, microbes from soil samples were cultured and their constituents extracted. This led to the isolation and identification of dozens of new classes of antibiotics, many of which were commercialized. Large numbers of these original agents, as well as chemical derivatives, are still used for the treatment of infectious disease (da Cunha et al., 2019). While the development of drug resistance has limited the continued use of many naturally occurring antibiotics, their discovery was the cornerstone for the development of the modern pharmaceutical industry after World War II. Semisynthetic analogues of microbial antibacterials are still being approved as new molecular entities (NME). As detailed in Table 1, a large proportion of natural product NMEs (7/26) were approved for antibacterial use in the period from 2008-2018.

Table 1.

Recent natural product drug introductions, 2008-2018.

Year Drug name Company Indication NP template
2008 Biolimus stent Biosensors Stenosis Rapamycin
Ceftobiprole medocaril Roche Antibacterial Cephalosporin
Methyl naltrexone bromide Wyeth/Progenis Constipation Morphinan
2009 Nalfurafine HCl Toray Pruritis Morphinan
Tebipenem pivoxil Spero Antibacterial Thienamycin
Televancin Theravance Antibacterial Vancomycin
2010 Cabazitaxel Sanofi-Aventis Cancer Taxane
Ceftaroline fosamil Takeda Antibacterial Cephalosporin
Eribulin mesylate Eisai Cancer Halichondrin
Fingolimod Novartis Multiple sclerosis Myricocin
Mifamurtide Takeda Cancer Muramyl dipeptide
Romidepsin Fujisawa/Astellas Cancer Cyclopeptide
Vinflunine ditartate Pierre Fabre Cancer Vinca alkaloid
Zucapsaicin E Merck AG Topical analgesic Capsaicin
2011 Fidaxomicin Optimer Antibacterial Macrolide
Spinosad ParaPro Antiparasitic Spinosyn A
2012 Arterolane Ranbaxy Antiparasitic Artemisinin
Carfilzomib Proteolix.Onyx Cancer Peptide
Crofelemer Salix/Shaman Diarrhea Oligomeric proanthocyanin
Dapagliflozin Astra Zeneca Diabetes Phlorizin
Ingenol mebutate Peplin/Leo Cancer Phorbol ester
Novolimus Elixir Stenosis Rapamycin
Omacetaxine mepsuccinate ChemGenex/ Teva Cancer Homoharringtonine
Pixanthone dimaleate Cell Therapeutics Cancer Anthracycline
2013 Canagliflozin Jannsen Diabetes Phlorizin
2014 ---
2015 Ceftolozame Astellas/Cubist Antibacterial Cephalosporin
Dalbavancin Vicuron/Durata Antibacterial Vancomycin
Empagliflozin Boehringer/Lilly Diabetes Phlorizin
Nalogexol Nektar/Astra Zeneca Consitpation Morphinan
Oritavancin Lilly/Medicines Co. Antibacterial Vancomycin
Tofoglozin Chugai/Sanofi/Kowa Diabetes Phlorizin
2016 ---
2017 ---
2018 Midostaurin Novartis Cancer Staurosporine
Naldimedine Shionagi Constipation Morphinan

1.3. Taxol

To stimulate interest in the development of cancer therapeutics, beginning in 1960 the US National Cancer Institute supported an extensive academic network examining plant sources for potential anti-cancer agents. Taxol (Wani et al., 1971) and camptothecin analogs (Wani and Wall, 1969, Lerchen, 2002) were the most prominent products that resulted from that program, although neither was commercialized until the early 1990s. Difficulty in obtaining the quantities of taxol needed for commercialization slowed its advancement, while the poor solubility of camptothecin required modifications to its chemical structure for it to have the physicochemical properties needed for clinical use. Once launched, taxol revolutionized the treatment of certain cancers and continues to play a major role in the management of these conditions.

2. Why has pharmaceutical company interest in natural products waned?

Over the past two decades research on natural products has declined in the pharmaceutical industry. Companies such as Merck (Mullin, 2008), Pfizer, and Bristol Myers Squibb have during this time closed in-house programs in natural products. This trend has been most visible in the United States, with a few European and Japanese companies continuing to support natural products groups. Several reasons have been given for this trend:

2.1. Discovery and development of natural products is perceived as a slow process.

This is a valid concern. Current high throughput screening (HTS) campaigns are designed to identify and prioritize chemical leads from compound libraries within weeks or months. If unpurified natural product extracts are tested initially, the time needed to isolate and identify active constituents make it impossible for natural product isolation to match the speed achieved by testing libraries of pure compounds. As detailed below, strategies have been developed to address this issue.

Natural product samples are most often tested as whole fermentation broths, or as crude extracts of plants and marine organisms. Once a hit is identified in a biological screen the extract must be fractionated to isolate the active compounds. This process typically requires that bioassays be conducted at each level of purification. Thus, the length of time required to conduct the bioassay and report the results, and the number of separation cycles needed to obtain pure compounds, are factors which dictate the time required to process a natural product hit. Even when cycles are performed on a weekly basis using a rapid bioassay, it is unusual for a natural product extract hit to yield a pure compound in less than a month. Other factors that may affect the speed and efficiency of compound identification are chemical instability, challenging separations, and between-run variation in bioassays.

2.2. All of the easy natural product drug discoveries have been made.

This perception is often reinforced by the expression “That pond’s all fished out.” However, while the number of plant and animal species is finite, only a very small fraction of all species has been chemically investigated, let alone broadly examined in a panel of bioassays. The number of higher plant species is estimated to be between 300,000 and 400,000. The largest plant screening program in pharma was conducted by Smith Kline & French, with about 19,000 species screened for alkaloid content using a simple color test (Raffauf, 1996). Over the past 20 years the U.S. National Cancer Institute has collected higher plants for screening, with the collection currently containing about 30,000 species, or roughly 10 percent of the total.

There is no simple way to tally the number of microbial samples that have been screened for biological activity, as the typical protocol in microbial screening is to perform only minimal species identification before undertaking tests for biological activity. Although the number of microbial samples screened is enormous, the taxonomic diversity of these samples is limited by the bias for collecting soil samples and the difficulty in growing all but a small fraction of microbes in culture. Recent advances in environmental microbiology have revealed that there is an enormous amount of unsampled microbiota (Epstein and López-Garcia, 2008, Tedersoo et al., 2014). Also, genomics studies have shown there is a correspondingly enormous amount of biosynthetic “dark matter” lurking in the uncultivated microbial world (Katz and Baltz, 2016). Thus, rather than the pond having been fished out, it appears that new types of bait or new fishing strategies are required to exploit fully the therapeutic potential of natural products. Heterologous expression of biosynthetic modules in host microbes amenable to manipulation is one approach developed for this purpose (Harvey et al., 2018), with activation of silent gene clusters being another (Ravindran, 2017).

Marine invertebrates were extensively studied over the past three decades yielding a great deal of information on the chemistry and biology of their bioactive constituents (Carroll et al., 2019). While the extent of biodiversity among marine invertebrates is unknown it is most likely quite large, given that life evolved first in the marine environment. The true diversity of marine life will not soon be known, at least by using classical methods as there are too few taxonomists to identify and classify new species, and only the easily SCUBA-accessible, shallow, warm marine waters have been extensively explored. Metagenomic methods are now being applied to marine invertebrates to address this issue (Lopez et al., 2019). Symbiosis between microbes and marine invertebrates is an important process that is often mediated by small molecules produced by the microbial partner(s) to defend the macro-organism partner (Florez et al., 2015).

The argument that there is little more to be discovered in natural products is reminiscent of the claim by some 19th century physicists that their field was nearing completion. While this was perhaps true for Newtonian physics, events of the last century have clearly demonstrated how misguided was their perception. Even if new developments in natural products consist of humble improvements in techniques and understanding rather than revolutionary advances in drug development, it is clear that many “fish” still remain in the pond.

2.3. The complex structures of natural products are too difficult to synthesize.

The chemical structures of natural products range from very simple (e.g., pinene, Figure 1a) to extremely complex. Improvements in structure elucidation techniques have made it possible to determine the complete stereostructure of natural compounds as complex as maitotoxin (Sasaki et al., 1996, Zheng et al., 1996), a compound with a molecular weight of 3425 Da incorporating 98 chiral centers (Figure 1b). Although such agents are likely to never be candidates for commercial scale total synthesis, the molecular weights of most of the natural products isolated and elucidated to date are <1000 Da, and would be reasonable candidates for commercial synthesis. In addition, many commercial drug products have been developed by synthetic modification of a naturally produced precursor. In that case total chemical synthesis is not required. Alternatively, structure-activity studies in conjunction with total synthesis may identify fragments of the parent structure with biological activity, thereby allowing for a drastic reduction in the size and chirality of a bioactive natural product. As discussed below, two examples where this approach has succeeded are bryostatin (Wender et al., 2017) and halichondrin (Yu et al., 2011).

Figure 1.

Figure 1.

Extremes of complexity in natural-product structures: (A ) Pinene and (B ) Maitotoxin.

2.4. Resupply of the source organism is difficult.

Obtaining the large amount of a natural compound needed for preclinical studies can be challenging. If derived from a plant which grows in a remote tropical location, physical access for recollection may be difficult, or permission to collect and ship the material difficult to obtain. Another risk is that the plant may only produce quantities of the desired compound under certain environmental or ecological conditions. A marine organism may require an expensive expedition, especially if the animal inhabits deep water or regions with strong or unpredictable currents. Even with a constant supply of material, the factors that induce production of the target substance may be poorly understood. This can result in a high degree of variability in the amount of active material harvested among batches. Understandably, pharmaceutical companies prefer predictable, controllable sources of starting material. Also, for commercial viability, solutions must be found to overcome the vagaries of natural product production. Approaches being taken to addressing these issues are covered in section 5, below.

2.6. High throughput chemistry is viewed as better than natural products.

Parallel synthesis techniques make possible the production of synthetic libraries containing thousands of distinct compounds. However, such rapid synthetic techniques have not significantly increased the rate of identification of drug candidates. Early combinatorial libraries were composed of compounds with poor solubility, with few useful hits found. In some cases, the quantities of compound produced were quite small and the purity highly variable. In recent years, smaller, more focused libraries have increased the yield of potential drug leads. Experience suggests that parallel synthesis is most useful for expanding an existing lead, rather than in creating screening libraries that yield NCEs of with therapeutic potential.

2.7. Natural product compositions of matter may not be patentable in the US.

Two US Supreme Court decisions have raised potential barriers to the patenting of natural products in the United States: Mayo v. Prometheus (Prometheus, 2011) and Association for Molecular Pathology v. Myriad (Myriad, 2013). Mayo concerned patenting a method of administering thiopurine drugs to treat autoimmune diseases which entailed the measurement of thiopurine metabolites to guide dosage of the parent drug to reduce toxicity. The Court struck down the patent because it felt the claims were made for underlying laws of nature, i.e., the variations in drug metabolism. In Myriad, the patents concerned the BRCA1 and BRCA2 tumor suppressor genes. Because mutations of these genes increase the risk of breast and ovarian cancer, it was argued these mutations would be useful diagnostic targets. The claims covered the naturally occurring gene and the DNA sequence, with the Court ruling the patents were similarly invalid. Both these decisions had immediate negative impacts on the patenting of diagnostics (Aceto, 2013, Dorn, 2013, Gordon, 2014).

With respect to natural products, in 2014 the US Patent and Trademark Office issued guidance to its patent examiners that interpreted the Mayo and Myriad cases as blocking composition of matter claims on isolated natural products (Anonymous, 2015, Lee, 2015). The unpopularity of the policy prompted revisions (Servick, 2015, Bahr, 2016, Lee, 2016) but these did not substantively change the position of natural products. A proposal for a legislative remedy (or a test case) was made (Tallmadge, 2017), and a draft bill was recently submitted in the US Senate (Servick, 2019). It remains to be seen how this issue will be resolved. No cases of rejection of patent applications or invalidation of existing composition of matter claims for natural product structures have yet occurred.

3. Why natural products are attractive for drug discovery

3.1. Secondary metabolites have evolved to be bioactive.

The metabolic energy and the genetic cost of the biosynthetic machinery to synthesize a small molecule requires that the compound provide some benefit to the organism, whether by defending it against predators, communicating with other members of its species, or interfering with competing organisms. While most functions of natural products in their producing organism are currently unknown, opinion has shifted markedly since the days when natural products were viewed as mere waste products (Mothes, 1969). Whatever the precise role, it appears now that, like drugs and hormones, many natural products interact with receptor sites on or within cells. The large number of pure natural products found to interact with particular mammalian receptors testifies to the inherent bioactivity of natural products. For example, natural product ligands known to interact with the mammalian GABA receptor include muscimol (Brehm et al., 1972), bicucculine (Johnston et al., 1972), securinine (Beutler et al., 1985), and picrotoxin (Akaike et al., 1985).

3.2. Structures are not limited by the chemist’s imagination.

While chemists may be as creative as natural systems, the natural systems have been at it for a much longer time. The most obvious value of natural products chemistry is the introduction of novel molecular skeletons and functionalities that have not previously been conceived of by humans. Some examples include the cephalostatins (Pettit et al., 1988), mitomycins (Stevens et al., 1965), and esperamicins (Golik et al., 1987), (Figures 2a, b, and c, respectively).

Figure 2.

Figure 2.

Structures of (A ) cephalostatin 1, (B ) mitomycin C, and (C ) esperamicin A1.

3.3. The Lipinski rules of five do not apply to natural products.

These rules were developed to drive synthetic chemists towards creating compounds that have the biophysical properties required of an orally active drug candidate. The rules require that compounds should have a molecular weight < 500 Da, posses <5 hydrogen bond donors, <10 hydrogen bond acceptors, and have log P<5 (Lipinski et al., 1997). Lipinski explicitly excluded natural products from these rules, primarily for the reasons set forth above (see Secondary metabolites have evolved to be bioactive), and because they often enter cells by way of membrane transporters rather than passive diffusion (Lipinski, Lombardo, Dominy and Feeney, 1997).

4. High throughput screening and natural products

4.1. Miniaturization and reductionism.

High throughput chemical screening to identify drug candidates evolved in the 1980s from automated clinical analyzer technology and miniaturization aimed at increasing the pace of testing and lowering the cost of testing per sample. As a result, robotic methods of sample manipulation and specialized detectors capable of reading 96-well microtiter plates were developed. At the same time the testing endpoint shifted from empirical measures of cell growth or function to specific interactions with protein molecular targets.

4.2. Cell-free or cellular assays?

In its most extreme reductionist forms, targeted screening involves detection of the interaction of test compounds with a purified, naked protein. Hits from that experimental model are then tested in a functional assay before progression to a cellular, and then tissue level of complexity. Because the highest level of reductionism provides the lowest barrier to successfully identifying hits, the large number of compounds that test positive must be winnowed using secondary, tertiary, and even quaternary assays. Abundant and common natural products such as tannins (7.2.1) overwhelmed reductionist assay strategies with very high hit rates.

There has been a substantial shift in the last decade the use of screening assays that measure biological function directly in cells. As these can typically be tuned to higher stringency, they yield lower hit rates, with positive samples more likely displaying the desired biological properties than is the case with less discriminating tests.

4.3. Change the assay or change the sample?

Natural products do not always behave as expected even in cellular assays, or in functional cell-free assays. The question arises as to whether it is better to adapt the assay to the sample or vice versa. Both approaches have had some success, with the path chosen depending on the availability of resources in chemistry and biology.

A common problem with screening natural product extracts is that many fluoresce in the fluorescein wavelength range (emission maximum 521 nm). This results in a high false positive rate in a screen with a fluorescent endpoint. If the fluorophore endpoint is changed to a label that emits at >560 nm (Cy3B, for example), much less sample autofluorescence is seen, and the false positive rate declines. Alternatively, use of a time-resolved fluorescence label also substantially decreases sample interference. Most sample autofluorescence has a short half-life (i.e., 10 ns), while europium fluorescence labels, for example, have a much longer half-life (ca. 700 ms). Thus, by gating the photodetector to record the signal after a 1 ms delay, the majority of the sample autofluorescence is filtered out, while the label is sensitively detected (Hemmilä and Webb, 1997).

4.4. Prefractionation of extracts

One approach to sample modification that has attracted significant interest is “prefractionating” the crude extract. In its most complex form, this means isolating pure compounds and partially characterizing them before they are tested in bioassays. Several companies have taken this approach, with mixed success (Bindseil et al., 2001, Eldridge et al., 2002). Simpler, lower cost strategies that separate the crude extract into 5-15 samples based on a single chromatography step, followed by solvent evaporation, may provide much of the benefit at a reduced cost (Bugni et al., 2008, Wagenaar, 2008). These approaches all require an investment in automation. Automated weighing capability, flexible programmable liquid handling, and low cost separation media are required to produce the samples and conduct the assays.

There are several benefits with this type of approach: 1) cytotoxic compounds which might mask the activity of another compound in a cellular assay may be separated; 2) minor constituents are concentrated and can be tested at higher concentrations; 3) very polar or lipophilic constituents of an extract can be ignored or discarded entirely. The initial testing results from using prefractionation strategies instead of crude extracts have demonstrated higher hit rates in cellular screening assays (Henrich et al., 2006, Ruocco et al., 2007, Booth et al., 2009, Blees et al., 2010), and higher reconfirmation rates in biochemical screens (Bermingham et al., 2017). The US National Cancer Institute recently instituted a program to prefractionate its complete natural product extract collection for distribution to external screening groups. A library of 150,000 samples is currently available, and the number of samples is projected to reach 1 million (Thornburg et al., 2018).

5. Sourcing

5.1. Natural product sources for drug discovery

As noted above, plants have historically played the leading role in providing drugs or templates for drugs, with microbes following as illustrated in the antibiotic era. Screeners began examining marine sources in earnest once SCUBA facilitated the collection of algae and marine invertebrates. While only a few marine natural products have yielded commercial drug products, many marine compounds have demonstrated activity in screens, with a number of them undergoing preclinical evaluation. Obtaining an adequate supply of compound has been a major hurdle in the study of marine invertebrate agents. For example, bryostatin 1 was initially obtained for clinical studies from its marine source organism, Bugula neritina, using Good Manufacturing Practices (Schaufelberger et al., 1991). However, a mere 18 g of material was purified from 14,000 kg of the producing bryozoan. Mariculture of the same animal was subsequently accomplished, leading to the successful production of large quantities of bryostatin 1 (Mendola, 2003).

A few programs have used insects as a screening source, such as in a collaboration between the Merck and InBio in Costa Rica (Sittenfeld et al., 1999), and in the Eisner lab at Cornell University (Schröder et al., 1998). Also notable is the work of John Daly using amphibians as a rich source of bioactive compounds (Daly et al., 2005). Epibatidine, a frog alkaloid (Badio and Daly, 1994), served as the stimulus for the design of the analgesic drug candidate ABT-594 (Arneric et al., 2007).

5.2. Microbial or dietary origin of marine and plant metabolites

Natural product investigators often encounter difficulties in obtaining reliable production of desired compounds from the producing organism. For example, it is common in microbial screening to confirm bioactivity by regrowing the microbe in the same conditions under which the initial screening sample was produced. In these cases, a success rate of only 50 percent is common. Similarly, when a plant is collected for isolation of larger amounts of constituents, it is not unusual to find low amounts of the desired metabolite, or no compound at all. This is even more common with marine invertebrates.

The reasons for these problems are not fully appreciated, but undoubtedly manifold. With microbes, obtaining good production of a desired metabolite is often a matter of studying the culture conditions (growth media, time, temperature, oxygenation, etc.) and defining the best conditions for reliable production. With plants, the chief problem may be a poor understanding of taxonomy. That is, careful botanical field studies may reveal several closely related species, only one of which produces the compound in question (McKee et al., 1998a). Dependence of metabolite production on environmental factors (climate, season, herbivore pressure) often plays an important role for plants. In marine invertebrates a poor understanding of taxonomy contributes to lack of reproducibility. Additionally, vectoring of metabolites from one organism to another and sequestration in the second organism is known to contribute to variability in production among samples (Thoms et al., 2006, Puglisi et al., 2019, Zan et al., 2019). Dietary sources of bioactive compounds have also been identified in amphibians that consume arthropods and other small leaf litter animals, along with the mechanisms of sequestration (Daly, Spande and Garraffo, 2005, Saporito et al., 2007, Caty et al., 2019) .

Another reason for erratic production may be that the higher organism is not the source of the compound which is instead produced by a microbial symbiont. Support of this comes from the fact that in many cases marine invertebrates have been found to contain compounds which look suspiciously like microbial metabolites (Simmons et al., 2008). Occasionally , similar compounds have been isolated from both a marine invertebrate and a microbe (Suzumura et al., 1997, McKee et al., 1998b). If the microbe is an obligate symbiont, it may be difficult to obtain proof of the relationship. A case has been made by Haygood’s group that bryostatins are produced by a symbiont, with the details of the symbiosis gradually being defined (Hildebrand et al., 2004, Miller et al., 2016). Similar reports for endophyte-plant derived compounds are also intriguing (Smith et al., 2008). An example is the isolation of taxol from an endophytic fungal associate of the Pacific yew and its antifungal utility to the plant host (Stierle et al., 1993, Soliman et al., 2015).

5.3. Synthesis of natural products versus biological production

Organic chemists have made great strides in being able to synthesize complex, chiral molecules such as natural products. While difficulty and cost are directly related to the number of chiral centers and molecular weight, total synthetic approaches for the production of natural products are becoming more common. Given sufficient resources, it is possible to reduce the number of synthetic steps required to reach the target molecule and improve the yield at each step, while using inexpensive starting materials. Wender’s group has developed synthetic routes to both bryostatin 1 (Figure 3a), and to “bryologs” (Figure 3b), compounds that display a potency similar to bryostatin 1 but with more simplified chemical structures. One recent, highly active bryolog, with differential potency between two PKC isoforms, was prepared in 10 steps in an overall yield of 30 percent (Wender, Hardman, Ho, Jeffreys, Maclaren, Quiroz, Ryckbosch, Shimizu, Sloane and Stevens, 2017).

Figure 3.

Figure 3.

Structures of (A ) bryostatin 1 and (B ) a bryolog.

Another example of synthetic success with a complex natural product is halichondrin B (Figure 4b). Wild collection of the producing sponge gave poor yields of the target compound (Uemura et al., 1985, Pettit et al., 1991). Mariculture in New Zealand yielded similar levels of halichondrin B (Munro et al., 1999). When a total synthesis was accomplished by the Kishi group (Aicher et al., 1992), it was discovered that fragments half the size of the natural product possess all of the bioactivity (Wang et al., 2000, Dabydeen et al., 2006). In 2010 Eisai gained FDA approval of eribulin (Figure 4b) for the clinical management of some types of cancer (Huyck et al., 2011).

Figure 4.

Figure 4.

Structures of (A ) halichondrin B and (B ) its simplified analog eribulin.

An example from my own group involves englerin A, whose structure was published in January 2009 (Ratnayake et al., 2009). This small sesquiterpene diester contains seven adjacent chiral centers, which constitutes a substantial challenge for total synthesis. Nonetheless, within a year, with no prior disclosure, the Christmann group reported its total synthesis (Willot et al., 2009). Many other groups followed with various efficient synthetic approaches, notably those of the Chain (Li et al., 2011) and Echavarren (Molawai et al., 2010) laboratories. Together all of the many studies have provided access to over 100 analogues in a relatively short time. Analogues of englerin A are currently in advanced preclinical development as antineoplastic agents (Wu et al., 2017).

5.4. Biosynthesis in heterologous organisms

Improvement in DNA sequencing tools have facilitated investigations of the biosynthetic pathways that lead to secondary natural products (Walsh, 2015). The biosynthesis of polyketide natural products has attracted a great deal of attention because many commercial antibiotics are derived from this pathway. However, non-ribosomal peptide synthesis, terpenoid biosynthesis and flavonoid pathways have also been elucidated in many organisms. A key observation is that many such pathways consist of modular gene clusters that can be manipulated as an entire unit, particularly in bacteria (Donadio et al., 1991). Polyketide synthase modules share enough homology that they can be identified in relatively distantly related organisms by lowering the stringency of hybridization reactions. While most biosynthetic gene clusters are not expressed under typical cultivation conditions, cryptic clusters can be turned on by a variety of elicitors to stimulate production (Xu et al., 2017).

In fact, such modules are detected in uncultivatable microbes (Piel, 2002), where elicitation is not an option. Metagenomic studies of environmental samples have become quite popular, with analysis of different environments yielding detailed information on the capabilities of organisms that cannot be cultivated by conventional techniques. For example, a recent study of northern California grassland soil reconstructed hundreds of nearly complete bacterial genomes containing a large repertoire of biosynthetic genes (Crits-Christoph et al., 2018).

This opens up the possibility of expressing the module in a convenient heterologous organism to obtain the desired secondary metabolite, assuming the needed precursors are available and other cellular machinery is compatible with the production of the metabolite (Zhang et al., 2008). This involves “refactoring” to adjust the suitability of the conditions and host organism to allow the desired gene cluster to be expressed in a functional state. One such platform has been described for fungal natural products (Harvey, Tang, Schlecht, Horecka, Fischer, Lin, Li, Naughton, Cherry, Miranda, Li, Chu, Hennessy, Vandova, Inglis, Aiyar, Steinmetz, Davis, Medema, Sattely, Khosla, St Onge, Tang and Hillenmeyer, 2018).

In addition, by altering the module, altered analogous metabolites may also be produced (Xu et al., 2009). It has even become common to predict the biosynthetic product from the sequence of a polyketide module (Banskota et al., 2006).

Progress with plant biosynthetic pathways has been slower than for microbes, primarily due to the scattered distribution of biosynthetic genes in the organism. The Keasling group recently succeeded in expressing the complete biosynthetic pathway for cannabinoids in yeast, (Luo et al., 2019), as well as a partial pathway to artemisinin in yeast, completed by chemical synthesis (Paddon et al., 2013). The Møller laboratory has made good progress on selected terpenoid plant systems inserted in yeast, including the concept of combinatorial biosynthesis (Møller and Ratcliffe, 2014, Andersen-Ranberg et al., 2016). These strategies were reviewed recently (Cravens et al., 2019).

5.5. Ethnobotany

Knowledge of the medical effects of plants is not limited to European science and cultural traditions. Botanists trained in anthropology have studied many non-western cultures to inventory their use of plants and other natural substances for medical and other purposes (Buenz et al., 2018). Chemical and pharmacologic investigation of ethnobotanical information is a viable alternate pathway to high throughput screening for drug discovery, although it has its own limitations.

First, cultural concepts of disease can vary. While most cultures readily recognize a superficial fungal infection or diabetes in a similar manner, most cancers are not diagnosed and interpreted in the same manner across cultures (Hartwell, 1967). Nonetheless, some have claimed that plants used traditionally for medicinal purposes yield a higher fraction of anti-cancer activity than unselected plants (Spjut, 2005). In addition, the medical effects of many plants used in traditional medicine may be less specific than what is desired by western pharmacologists. For example, while tannins are often found in herbal preparations and may contribute to their biological activity, tannins are usually not well-suited to drug development. Nonetheless, SP-303, developed by Shaman Pharmaceuticals, (Holodniy et al., 1999) was a carefully defined tannin preparation from Croton lechleri, a Peruvian ethnobotanical (Williams, 2001) that was tested for several antiviral indications before being sold an over-the-counter anti-diarrheal agent. In 2013 it entered the prescription market as Crofelemer (Chordia and MacArthur, 2013).

Another issue is exemplified by both Chinese traditional medicine and the Indian Ayurvedic system. Both of these ancient traditions utilize polyherbal preparations for the majority of prescriptions. Each component is thought to play a particular role, in some cases by modulating the toxicity of another component. This complexity makes active principle analysis difficult. Reductionist approaches to Chinese and Ayurvedic preparations have been largely unsuccessful in validating their traditional uses, although many bioactive molecules have been isolated from both (Tang and Eisenbrand, 1992, Deocaris et al., 2008). The use of microarrays to study the in vivo effects of complex preparations may hold some promise for better understanding of these products and there future clinical use (Yin et al., 2004).

5.6. Chemical ecology

While natural products are now known to not be waste products of the producing organism, (Mothes, 1969) the purpose they serve for the producer is rarely similar to their potential use in human medicine. Most drugs act through interaction with protein receptors. Domains of proteins, though not their precise function, are widely conserved (Rompler et al., 2007). Thus, ligands targeted to a particular domain in nature may also have activity in an orthologous or paralogous receptor in humans. C. elegans, for example, has been proposed as a model organism for anti-Parkinson drug screening; many of the compounds which affect dopaminergic systems in humans also have more or less parallel effects in worms (Nass et al., 2008).

Investigation of the ecological function of natural products is a field unto itself, and elucidation of the role a compound plays can be experimentally challenging. Those that have been identified include antifeedant effects (Lidert et al., 1987), allelopathy (interference with growth of competitors) (Tseng et al., 2001), and endocrine disruption (Dinan and Lafont, 2006).

5.7. NCI Letter of Collection

In the late 1980s, contracts were developed by the U.S. National Cancer Institute for the collection of large numbers of plant, microbial and marine samples worldwide. The collectors required permits to collect in many different countries and those countries needed assurance that their rights would be respected in the drug development process. To that end, the NCI developed a standard Letter of Collection which is signed by both parties (Cragg and Newman, 2005). This letter states the willingness of the NCI to collaborate with source country scientists, to deposit voucher specimens in source country repositories, and to develop benefit-sharing arrangements when patents were filed. In addition, Memoranda of Understanding could also be developed to frame direct collaborations.

5.8. Convention on biodiversity

The NCI agreements predated and presaged the 1992 Rio Convention on Biological Diversity (CBD). While the U.S., alone among United Nations member states, has ratified but not signed the treaty, U.S. Department of State policy requires following its principles. The CBD calls for preservation of biological diversity, for protection from exploitation of source country genetic resources, for equitable sharing of the benefits of technology, and for technology transfer to the source country. The 2010 Nagoya Protocol focused on implementation of equitable benefit sharing under the CBD (Cragg et al., 2012, Joppa et al., 2013).

While it is generally perceived that the CBD made access to natural products resources more difficult, it has reduced the worst abuses of source countries by the developed world. It has not resolved the political issue of how benefits should be distributed within the source country, however. See for example the case of Hoodia, a weight loss product from the San people in South Africa where active constituents were patented by government scientists at the South African CSIR and licensed to Phytopharm plc and Unilever (Wynberg, 2004, Anonymous, 2006, Bladt and Wagner, 2007). Currently Hoodia products are only available as dietary supplements.

6. Techniques in natural products drug discovery

6.1. Extraction

Before tissues of an organism can be tested, they must undergo an initial extraction to separate the desired small molecules from the biopolymers (proteins, cellulose, chitin, nucleic acids) that make up the bulk of the material. For plants, before extraction it is common to dry plant parts thoroughly in the field at the point of collection, so the material does not decompose en route to the laboratory. To accelerate extraction, the dry tissue is ground using any of several mills (e.g., a Wiley mill, or a hammer mill). Alternatively, tissues may be frozen, although this is often expensive and cumbersome. Frozen material may be lyophilized. If DNA or mRNA is sought for cloning of proteins, flash freezing the freshly collected tissue in liquid nitrogen is required to obtain useful material.

There are very few standard techniques for extraction, because choice of solvent and conditions depends on the spectrum of small molecules desired. For extraction of drug-like molecules of intermediate polarity, the NCI has found percolation at room temperature with a 1:1 v/v mixture of dichloromethane and methanol to be useful (McCloud, 2010). Extraction techniques that involve heating the solvent and extracted compounds, as in a Soxhlet apparatus, are generally avoided unless the desired compounds are known to be heat stable. Otherwise, heating should be avoided in preparing samples of unknown composition for biological screening.

Marine invertebrate tissues present unique extraction problems because of high water and salt content. An approach adopted at the NCI has proven generally applicable for a wide variety of marine specimens. Frozen samples are broken into pieces small enough to be fed into a commercial hamburger grinder with CO2 pellets. The resulting powdered material is stored frozen long enough for the CO2 to sublime, then thawed briefly and stirred with water as a slurry. Filtration through paper in a low-speed centrifuge removes the mucilaginous tissue, and the resulting aqueous extract is freeze dried. The marc (remaining solid residue) is also lyophilized and then extracted with the methylene chloride-methanol mixture (McCloud, 2010).

The solvent must be removed from the solutions which result from any of these extraction procedures. This is done to make it possible to obtain a weight for the extracted material and to avoid chemical reactions in solution which may alter the constituents. Aqueous solutions are lyophilized, while organic solvent mixtures are dried using rotary evaporators. A final finishing under high vacuum removes most traces of the solvent. Materials should be stored in borosilicate glass bottles or vials at −20° C to ensure stability.

For high throughput screening applications, it is common to store libraries in dimethyl sulfoxide (DMSO). DMSO is an extraordinarily good solvent for most natural product samples, including extracts. Organic extracts can often be entirely dissolved in DMSO at concentrations of 10-100 mg/ml, while 50% DMSO solutions of aqueous extracts are possible. It should be noted that DMSO concentrations >25% generally suppress bacterial growth in solutions of aqueous extracts. The bulk extract material should not be stored in DMSO, however, since this solvent can facilitate a number of oxidation reactions. In addition, because of its hygroscopic nature DMSO causes moisture absorption even in nominally sealed microplates in the freezer (Ellson et al., 2005). Such extract plates should be reconstituted from bulk stocks on a regular basis to avoid sample deterioration.

Each bioassay will have a limit on the amount of DMSO it can tolerate. Typically this is 0.5-1% of assay volume for cellular assays. For biochemical assays, it is often as high as 5-10% of assay volume. The limit should be established in advance for a particular test and DMSO controls run in every experiment.

6.2. Separations

Once an extract has been confirmed as a hit in a biological assay, the active compounds in the extract must be identified. This is accomplished using an iterative process termed bioassay-guided fractionation. To this end, an extract is separated into several fractions and the parent extract and fractions tested in the assay. Several outcomes are possible. One is that all activity may be lost in the daughter fractions, in which case the separation method is deemed unsuitable. Loss of biological activity may be due to irreversible binding to the separation media, or to instability of the active compound. A second possibility is for all or most daughter fractions to display some low level of activity. This too is undesirable and is taken to indicate that the separation mode is unsuitable. The third and desired outcome is where one or several daughter fractions contain substantial amounts of bioactivity, with the mass of active fractions reduced from the parent with a corresponding increase in potency. A useful technique in monitoring separations is to calculate both mass and activity recoveries. Thus, if a 5 g sample of a parent extract is separated and yields a summed fraction mass of 4.5 g, the mass recovery is 90 percent. Using concentration-response curve data from the assay, bioactivity recovery is calculated by the equation

(MiIi)MpIp Equation 1

where Mi are the masses of the fractions, Ii are the IC50 values for each fraction, and Mp and Ip the respective values for the parent extract. If a fraction displays no activity, the term can be ignored. This calculation is limited by the precision of the bioassay, but can be useful in judging the success of a trial separation.

The invocation of synergism to explain loss of activity on fractionation has only rarely been proven experimentally. If activity is lost, most commonly it is attributed to compound instability or irreversible binding to chromatography media. Given a suitably precise assay, calculation of mass and activity recovery will often provide clues to the source of the problem.

A single separation step is rarely sufficient to obtain purified active compounds. While use of high performance chromatography can often yield a superb separation of complex materials, it is more cost-effective to save the high performance step for last because crude extracts can damage expensive preparative HPLC columns. The most useful first separation process is one based on polarity. For example, the so-called Kupchan partition employs a series of two-phase mixtures in a separatory funnel to sort components by partition coefficient. While simple, the technique suffers from a tendency to form emulsions, and from difficulty in evaporating to dryness the water-saturated organic layers. A more convenient approach for organic plant extracts involves solid phase extraction with diol bonded phase media, with increasingly polar solvents used to elute successive fractions (Beutler et al., 1990). This procedure can be scaled over a wide range of volumes, and introduces no water into the samples. For marine samples, a wide-pore C4 bonded phase scheme can be employed with methanol-water mixtures to separate the large amount of salts and other polar material from the more drug-like intermediate polarity fractions (Cardellina et al., 1993).

Intermediate resolution techniques such as flash chromatography, or gel permeation chromatography are useful once the polarity cuts are made. Open column systems using Sephadex LH-20 with a variety of solvents separate based on both size exclusion and adsorption mechanisms, can be very useful.

Final purification is most often accomplished by preparative HPLC. A wide variety of bonded phases are available (e.g., cyano, C18, phenyl, diol, amino,) which can be operated in reversed-phase or normal phase modes, as well as by ion exchange or hydrophilic interaction chromatography. Pilot thin layer chromatography experiments can provide useful hints on the best choice of column packing and elution conditions. Ultimately, analytical scale HPLC is used to define precise flow and solvent strength parameters. Even with relatively purified fractions, it is often useful to employ gradient elution to obtain an optimum separation. While C18 bonded phases dominate the analytical chemistry market, they are only one of the tools in the HPLC column drawer of a natural products isolation laboratory.

It is also important to carefully consider peak detection. It is common analytical practice to use UV detection at 254 nm, which is useful for compounds with suitable chromophores. However, many constituents of natural materials lack absorbance in this range. The most effective strategy is to use lower wavelengths for detection. For acetonitrile-water systems it is possible to use wavelengths as low as 200 nm to observe compounds with poor UV absorbance. Alternative methods are evaporative light scattering detection or refractive index detection. However, neither of these approaches are very well suited for larger scale separations.

Next, the separation must be scaled up to semi-preparative or preparative scale using larger diameter HPLC columns with the same length, column chemistry, particle size, and porosity used for the smaller samples. Loading studies with increasing injections of material establish the amount of mass than can be effectively separated in a single run. The high cost of larger columns is offset by the shorter time required to run the separation, with columns as large as 41 mm diameter being used with laboratory scale pumping systems capable of delivering to the column 50-100 ml/min of solvent. If flow rates and injection volumes are scaled proportionately, preparative separations can be obtained with the same reproducibility and resolution as analytical separations. The sample injected on an expensive preparative scale column must be carefully filtered and the solvent conditions chosen to elute virtually all of the applied sample. Otherwise, particles and other uneluted material will rapidly degrade column performance.

An excellent overview of preparative chromatographic techniques applied to natural product isolation is available in book form (Hostettmann et al., 1998) and in a recent review (Bucar et al., 2013).

6.4. Structure elucidation

Once the active compounds are obtained in pure form, they are subjected to structure elucidation. The key technique for this is NMR, specifically a series of two-dimensional experiments (COSY, HSQC, HMBC, NOESY) that make it possible to establish the connectivity of all hydrogen and carbon atoms. Serving a very important complementary role is high resolution mass spectrometry (MS), which is capable of providing precise mass measurements that identify the molecular formula of the compound. It is often possible to identify the structure of an unknown molecule using only NMR and MS. Other spectroscopic techniques, such as UV, IR, circular dichroism (ECD) and optical rotation, serve ancillary roles, although they may be critical in specific cases. As the number of atoms in a molecule increases, structure elucidation becomes more difficult because of the exponential increase in possible structures for a given formula. It is routine to determine structures of compounds under a molecular weight of 500 Da, while those over 2,000 Da nearly always require extensive chemical degradation or transformations to establish their structures. Exceptions are smaller biopolymers such as peptides which can be easily sequenced if all of the constituent repeating components are known.

The ability of NMR and MS to provide useful information from smaller amounts of compound has increased substantially in recent years. In particular, advances in NMR probe design, especially gradient probes, flow probes and cryoprobes, have greatly increased sensitivity (Reynolds and Enriquez, 2002). Higher field strength magnets have increased NMR spectral dispersion so that more peaks can be resolved in a spectrum. Improved NMR pulse sequences have reduced experiment time and increased resolution. One advancement is the construction of pulse “supersequences” which chain multiple 2D sequences in a single measurement, dramatically reducing acquisition time (Kupče and Claridge, 2017). Another is the use of residual chemical shift anisotropy in proton-poor molecules (Liu et al., 2018, Milanowski et al., 2018).

Similar improvements have been made in MS, with electrospray ionization and matrix assisted laser desorption being two ionization techniques that have proven to be highly valuable in natural product characterization. Some cutting edge techniques, such as Fourier transform cyclotron resonance mass spectrometry (FTICR-MS), are used mainly in industrial settings because of costs (Feng and Siegel, 2007).

Computational prediction of spectroscopic properties such as 13C-NMR shifts and ECD curves, has become more common in structure elucidation, particularly in cases where stereochemical features are difficult to define (El-Elimat et al., 2015, Tran et al., 2019).

An alternative technique for structure elucidation is x-ray crystallography, which has for decades been employed in natural product structure elucidation. It remains an important tool, especially for determining the absolute configuration of complex chiral molecules. The obvious limitation of this approach is that the compound of interest must be prepared in a crystalline form. If crystallization using conventional techniques is not possible, the compound can be derivatized with a variety of modifiers in an attempt to improve its ability to form crystals. Application of robotics to automatically generate many small-scale crystallization experiments has increased the likelihood of identifying crystallization conditions.

6.5. Crystalline sponges

A new development in crystallography is the “crystal sponge method” developed by the Fujita group at the University of Tokyo (Inokuma et al., 2013). As noted above, many natural products fail to crystallize using standard protocols. With host-guest technology the analyte is soaked into a porous metal-organic complex which is itself highly crystalline. If the analyte binds to the metal complex in a consistent orientation, x-ray diffraction studies can then be conducted on a very small scale. The Clardy group has explored the process to determine its scope and transferability and generated some guidelines for success (Ramadhar et al., 2015). Moreover, Fujita has updated the methodology (Hoshino et al., 2016). To date, most of the applications of this method for characterizing natural product structures were performed by the Fujita laboratory (Yoshioka et al., 2015, Matsuda et al., 2016, Urban et al., 2016, Brkljaca et al., 2017, Lee et al., 2017, Kai et al., 2018), although a Chinese group has published the absolute configuration determination of a plant metabolite, asarinin, using crystalline sponge methods (Li et al., 2019). It has been reported that crystalline sponges could be used as an affinity matrix to trap sesquiterpenes from a crude red algal extract for analysis (Wada et al., 2018).

6.6. Microelectron diffraction

A newer technique derived from cryoelectron microscopy has been developed that utilizes electron diffraction rather than x-rays to determine the structure of small molecules. Cryogenic cooling is important in reducing damage to the sample due to high energy electrons. Two groups published this procedure in 2018 describing their success in imaging very small crystals (~100 nm) of small molecules with a cryoelectron microscope (Gruene et al., 2018, Jones et al., 2018). Tiny crystals have the advantage of forming much more readily and with fewer defects than macroscopic crystals. While cryoelectron microscopes are expensive (~$6 million) and heavily booked for single particle studies of proteins and protein complexes, microED can be efficiently performed on equipment no more expensive than an NMR. The scope and difficulty of microED remain to be determined (Brown and Clardy, 2018, Nannenga and Gonen, 2019).

While the ability to determine novel structures with spectroscopic and crystallographic methods with smaller samples are important advances, animal testing has not been miniaturized. Therefore, it is always necessary to conduct preparative separations to obtain sufficient material for in vivo analysis.

6.7. Hyphenated techniques

Hyphenated techniques such as HPLC-MS, HPLC-UV, and HPLC-NMR are useful analytical platforms for detection, identification, and quantification of compounds in extracts. These serve as important tools for determining the compounds in a sample and may inform preparative separation methods. They also are important techniques for chemical dereplication (see below). In addition to coupling several different detection methods, HTS bioassays may be conducted on individual fractions to complement the physicochemical data. One of the drawbacks of using hyphenated techniques is the large data sets generated from each run. Managing, analyzing, and interpreting the results can be daunting.

7. Dereplication

7.1. Biological and chemical

With over 300,000 known small molecules characterized from natural sources, it is no surprise that previously known natural products are often re-isolated during bioassay-guided fractionation. While this may be a welcome discovery if the biological activity is new, a great deal of time and effort can be spent in de novo structure elucidation of known compounds. This problem first emerged during the search for new antibiotics, where the taxonomy of microbial cultures was generally not identified prior to screening. Efforts to avoid investing resources in the elucidation of known compounds are identified by the term dereplication (Corley and Durley, 1994). In all its forms, dereplication is based on attempts to shift the identification of known compounds to an earlier point in the discovery process, either before a pure active substance is isolated, or before a complete NMR data set is acquired and analyzed. Direct physical comparison with standard compounds is a very effective tactic. However, amassing a library of known compounds is an unattainable task for most laboratories.

Most effective is a combination of biological and chemical methods. If the source organism has been identified, reference to databases of known compounds such as the CRC Press Dictionary of Natural Products (Buckingham and Thompson, 1997), can suggest candidate structures. Physicochemical data, in particular ultraviolet spectra and mass spectra, can rapidly limit the scope of possible compounds, especially when combined with analytical HPLC (Lang et al., 2008). Efforts to construct a large public database of mass spectral data are beginning to bear fruit (Allard et al., 2016, Wang et al., 2016, Covington et al., 2017, Olivon et al., 2017, Nothias et al., 2018, Olivon et al., 2019). While NMR database tools have also advanced they are less comprehensive than the mass spectral findings (Bakiri et al., 2017, Zhang et al., 2017, Buedenbender et al., 2018). An effort to build a comprehensive NMR dataset of natural products has been proposed (McAlpine et al., 2019). A system capable of merging mass spectral and NMR data would be ideal (Zani and Carroll, 2017).

7.2. Nuisance compounds

Not all compounds contained in a natural product extract are viable as drug candidates. Several classes of such undesirables are described below.

7.2.1. Tannins

Tannins are polyphenolic plant metabolites that were initially discovered as the compounds responsible for tanning leather. Oak bark and many other plant materials contain substantial quantities of tannins, complex molecular structures which incorporate gallate esters (hydrolysable tannins, e.g. Fig. 5a) or flavanol polymers (condensed tannins, e.g. Fig. 5b) (Khanbabaee and Ree, 2001). Phlorotannins are a third class found in brown algae. Tannins play an important ecological role in deterring feeding by herbivores, and may be produced in response to tissue injury. Many tannins are reported to be antinutritional in that they reduce the digestible protein in foods (Butler, 1992). The mechanism for both the tanning effect and the antifeedant/antinutritional roles is related to noncovalent binding to proteins. Because this effect is a relatively nonspecific effect, with a given tannin being capable of binding to many different proteins, tannins are considered poor candidates for drug development. Much effort has been expended over the years in removing tannins from natural product screening samples because they can be active in a wide variety of cell-free and cell-based assays (Cardellina, Munro, Fuller, Manfredi, McKee, Tischler, Bokesch, Gustafson, Beutler and Boyd, 1993, Wall et al., 1996).

Figure 5.

Figure 5.

Figure 5.

Structures of some common nuisance compounds. (A ) A common condensed tannin, proanthocyanidin C1. (B ) A hydrolyzable tannin. (C ) A phorbol ester, phorbol 12-tigliate 13-decanoate. (D ) A saponin, ginsenoside Rb2. (E ) General repeating structure of a marine anionic polysaccharide. (F ) General repeating structure of a cationic polymeric alkylpyridine, halitoxin.

7.2.2. Phorbol esters

Phorbol esters are diterpenes produced exclusively by plants in the Euphorbiaceae and Thymelaeaceae families (e.g. Fig. 5c). Many compounds of the class are skin irritants and tumor promoters. Their mechanism of action is activation of intracellular protein kinase C (PKC) (Nishizuka, 1984). Because many cellular functions are PKC-dependant, phorbol esters are pleiotropic agents that affect many cellular pathways. Hence, although phorbol esters appear as hits in a number of cellular screens, they are not good drug candidates because of their potential toxicities and tumor-promoting properties. The general distribution of phorbol esters in different species has been described (Beutler et al., 1989, Beutler et al., 1990, Beutler et al., 1995, Beutler et al., 1996).

7.2.3. Saponins

Saponins are glycosides of triterpenes or sterols produced by many plants (Hostettmann and Marston, 1995). The number of sugar residue varies from one to a dozen, and other chemical functionalities may be appended in various ways (e.g. Fig. 5d). Their ability to act as detergents and form foams in water solution is related to their use as soaps and for killing fish. In the context of biomedical screening assays these properties lead to cell lysis, which can be yield either a false positive response or to interference, depending on the nature of the assay endpoint. In addition, some saponins cause hemolysis, an undesirable property for a drug candidate. A diagnostic feature of saponins in a cell growth assay is that cell lysis is an extremely rapid process, occurring over several minutes, whereas other cell-killing mechanisms generally require numbers of hours. Thus, time-course studies are used to help to distinguish saponins from other types of hits. It is important to note that because not all saponins are detergents or hemolytic some may be viable drug leads (Bento et al., 2003, Tang et al., 2007).

7.2.4. Anionic polysaccharides.

Cellulose, a neutral polysaccharide, is the primary structural material of plant tissues. For animals, cartilage, which is composed of collagen and proteoglycan, plays a similar role in animal tissues. The carbohydrate portion of proteoglycan is composed of heavily sulfated N-acetylglucosamine and hexuronic acid units (e.g. Fig. 5e). These materials, which are often found in marine invertebrate aqueous extracts, are of high molecular weight and carry a substantial negative charge (Beutler et al., 1993). Although anionic polysaccharides are highly active in cellular HIV assays (Beutler, McKee, Fuller, Tischler, Cardellina, Snader, McCloud and Boyd, 1993), their high molecular weight and heterogeneity make them unattractive as drug candidates. Sulfated cyclodextrins, which have been studied as potential antiviral agents, have substantially the same antiviral activity as anionic polysaccharides without some of their liabilities (Moriya et al., 1993). Sulfated polysaccharides are encountered as hits in a variety of cellular screens. They may be removed from extracts by precipitation from ethanol solution at low temperatures. Plants also produce anionic polysaccharides, although they generally display weaker activity than those found in animal tissues.

7.2.5. Cationic polymeric alkylpyridines.

Less often, cationic polymers are found as nonselective hits from natural product extracts. Marine sponges are the usual source of these compounds (e.g. Fig. 4f) (Schmitz et al., 1978, Davies-Coleman et al., 1993).

7.3. Pattern matching

Another approach to identifying or eliminating known natural products without investing resources in their re-isolation and characterization is to compare biological and chemical “fingerprints” to standards. By using the results of multiple biological and chromatographic experiments in which the standard compounds have previously been tested, it is possible group similar samples together, and to propose a dereplication hypothesis for the samples whose results match those of a known compound.

The most data-rich environment in which this has been done is for the NCI 60-cell results. Because thousands of natural products have been tested, these can be used as reference points in data analysis in comparison with results from fractions of crude extracts. A variety of mathematical approaches have been used for the analysis, including calculation of Pearson correlation coefficients (Paull et al., 1989), neural networks (Weinstein et al., 1992), and self-organizing maps (Keskin et al., 2000). If the mechanism of action of the reference compound is known, the correlated test samples can be rapidly screened to determine whether they display similar mechanisms (Paull et al., 1992, Weinstein et al., 1997). This approach has been utilized for agents affecting tubulin (Paull, Lin, Malspeis and Hamel, 1992), epidermal growth factor pathways (Wosikowski et al., 1997), and vacuolar ATPase inhibitors (Boyd et al., 2001), among others.

There is no a priori reason why pattern matching must be limited to cell growth inhibition data. In fact, any type of data can, in principle, be mixed, even chromatographic, spectroscopic and taxonomic information. The utility of pattern matching depends mainly on the number of dimensions present in the data matrix. While redundant dimensions (i.e., cells which respond identically) do not contribute, scattered missing data are only a minor issue if the appropriate analytical techniques are applied.

8. Recent natural product drug introductions

Natural products and their derivatives continue to be approved as new drugs (Table 1). The information on Table 1 is not comprehensive as it excludes peptide drugs, antibody-drug conjugates and other agents that could arguably be considered natural product derivatives. For more comprehensive discussions of approved natural product drugs or those undergoing clinical testing, see the reviews by Cragg and Newman, and Butler (Butler et al., 2014, Newman and Cragg, 2016).

9. Which companies are still conducting natural products discovery ?

While natural products groups have been eliminated in most large pharmaceutical companies in the U.S., they remain active in Europe and Japan. Of the companies listed in Table 1, Bristol Myers Squibb, Merck, Johnson & Johnson, Pfizer, Glaxo Smith Kline, and Lilly no longer maintain in-house natural product discovery groups. Up until its merger with Pfizer, Wyeth had such a group at its Pearl River facility, which has since been closed. In Europe, Novartis continues its efforts in natural product drug discovery.

A corresponding trend is development of smaller, boutique companies that specialize in natural product drug discovery with the aim of licensing their leads to larger entities for clinical development and marketing (Gullo and Hughes, 2005). Pharmamar is an example of a small company that has had success in bringing a natural product (Yondelis) to market in collaboration with a major pharmaceutical firm. Nereus Pharmaceuticals advanced a marine microbial proteasome inhibitor (NP-0052, Marizomib) into phase Ib combination trials in cancer. Triphase purchased the rights to this agent and is conducting phase III trials in partnership with Celgene (ClinicalTrials.gov). Kosan Biosciences, which developed epothilone analogs using biosynthetic technology, was acquired in 2008 by Bristol Myers Squibb on the strength of its development pipeline, with ixabepilone being approved for cancer therapy (Cianfrocca, 2008). Small companies can also serve as screening contractors or provide the natural product libraries and expertise for pharma screening (e.g., Albany Molecular Research).

The public sector has taken on a larger role in drug discovery generally, and natural products drug discovery in particular. For example, an NIH-organized consortium has crowd-sourced 346 pure natural product screening samples from 45 primarily academic groups and screened the pilot set in 50 HTS assays (Kearney et al., 2018).

It is clear that the landscape of natural product research and drug development is rapidly changing. It is a major challenge to maintain the natural product knowledge base and resources developed by large companies. These resources are often lost with corporate mergers, acquisitions and restructuring. Exceptions include the preservation of the Merck natural products libraries at two locations (the Fundación Medina, in Spain, and the Natural Products Discovery Institute, in the US), and the Pfizer microbial culture collection donated to the Scripps Research Institute.

10. Diversity-oriented synthesis

The objective of diversity-oriented synthesis is to take its structural cues from nature. As a daughter of combinatorial chemistry, it seeks to meld parallel synthesis with chiral synthesis technologies. Thus, natural product scaffolds are designated as privileged structures and then functionalized by parallel synthesis (Balthaser et al., 2011, Smith and Kim, 2011, O’ Connor et al., 2012, Garcia et al., 2016). While attractive in concept, for the same reasons that natural products are desirable for drug leads (Section 3, above), the efficiency of this strategy remains to be determined. After all, functionalization choices are still made by the chemist.

11. Specialist journals in natural product science

Shown on Table 2 is a listing of specialist journals popular among those engaged in natural products research.

Table 2.

Specialist journals in natural products research.

Journal Name Publisher
Economic Botany NY Botanical Garden Press
Fitoterapia Elsevier
Journal of Antibiotics Japan Antibiotics Research Association/Nature
Journal of Chemical Ecology Springer
Journal of Ethnopharmacology Elsevier
Journal of Natural Products American Society of Pharmacognosy/American Chemical Society
Marine Drugs Molecular Diversity Preservation International
Molecules Molecular Diversity Preservation International
Natural Product Reports Royal Society of Chemistry
Natural Product Research Taylor & Francis
Pharmaceutical Biology Taylor & Francis
Phytochemical Analysis Wiley Interscience
Phytochemistry Elsevier
Phytochemistry Reviews Springer
Phytomedicine Elsevier
Phytotherapy Research Wiley Interscience
Planta Medica Thieme
Toxicon Elsevier

Acknowledgements

This research was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations implyendorsement by the U.S. Government.

References Cited

  1. Aceto JF 2013. Patent portfolios after Myriad, how to fit in those new genes? ACS Med. Chem. Lett 4:681–683. doi: 10.1021/ml400254f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aicher TD, Buszek KR, Fang FG, Forsyth CJ, Jung SH, Kishi Y, Matelich MC, Scola PM, Spero DM and Yoon SK 1992. Total synthesis of halichondrin B and norhalichondrin B. J. Am. Chem. Soc 114:3162–3164. doi: 10.1021/ja00034a086. [DOI] [Google Scholar]
  3. Akaike N, Hattori K, Oomura Y and Carpenter DO 1985. Bicuculline and picrotoxin Block γ-aminobutyric acid-gated Cl conductance by different mechanisms. Experientia 41:70–71. doi: 0. [DOI] [PubMed] [Google Scholar]
  4. Allard PM, Peresse T, Bisson J, Gindro K, Marcourt L, Pham VC, Roussi F, Litaudon M and Wolfender JL 2016. Integration of molecular networking and in-silico MS/MS fragmentation for natural products dereplication. Anal. Chem 88:3317–3323. doi: 10.1021/acs.analchem.5b04804. [DOI] [PubMed] [Google Scholar]
  5. Andersen-Ranberg J, Kongstad KT, Nielsen MT, Jensen NB, Pateraki I, Bach SS, Hamberger B, Zerbe P, Staerk D, Bohlmann J, Møller BL and Hamberger B 2016. Expanding the landscape of diterpene structural diversity through stereochemically controlled combinatorial biosynthesis. Angew Chem Int Ed Engl 55:2142–2146. doi: 10.1002/anie.201510650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Anonymous 2015. July 2015 Update: Subject Matter Eligibility, U.S. Patent and Trademark Office. [Google Scholar]
  7. Anonymous 2006. Protecting traditional knowledge: The San and Hoodia. Bull. World Health Org 84:345–345. doi: /S0042-96862006000500008. [PMC free article] [PubMed] [Google Scholar]
  8. Arneric SP, Holladay M and Williams M 2007. Neuronal nicotinic receptors: a perspective on two decades of drug discovery research. Biochem. Pharmacol 74:1092–1101. doi: 10.1016/j.bcp.2007.06.033. [DOI] [PubMed] [Google Scholar]
  9. Badio B and Daly JW 1994. Epibatidine, a potent analgesic and nicotinic agonist. Mol. Pharmacol 45:563–569. doi: 0. [PubMed] [Google Scholar]
  10. Bahr RW 2016. Formulating a subject matter eligibility rejection and evaluating the applicant’s response to a subject matter eligibility rejection, U.S. Patent and Trademark Office. [Google Scholar]
  11. Bakiri A, Hubert J, Reynaud R, Lanthony S, Harakat D, Renault JH and Nuzillard JM 2017. Computer-aided (13)C NMR chemical profiling of crude natural extracts without fractionation. J Nat Prod 80:1387–1396. doi: 10.1021/acs.jnatprod.6b01063. [DOI] [PubMed] [Google Scholar]
  12. Balthaser BR, Maloney MC, Beeler AB, Porco JA Jr. and Snyder JK 2011. Remodelling of the natural product fumagillol employing a reaction discovery approach. Nat.Chem 3:969–973. doi: 10.1038/nchem.1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Banskota AH, McAlpine JB, Sorensen D, Ibrahim A, Aouidate M, Piraee M, Alarco AM, Farnet CM and Zazopoulos E 2006. Genomic analyses lead to novel secondary metabolites. Part 3. ECO-0501, a novel antibacterial of a new class. J. Antibiot 59:533–542. doi: 10.1038/ja.2006.74. [DOI] [PubMed] [Google Scholar]
  14. Bento ES, Sant’Ana AEG, Hawkes GE, Calixto JB and Yunes RA 2003. The structure of velutinoside A: A pregnane pentasaccharide from Mandevilla velutina. Tet.Lett 44:8335–8337. doi: 10.1016/j.tetlet.2003.09.028. [DOI] [Google Scholar]
  15. Bermingham A, Price E, Marchand C, Chergui A, Naumova A, Whitson EL, Krumpe LRH, Goncharova EI, Evans JR, McKee TC, Henrich CJ, Pommier Y and O’Keefe BR 2017. Identification of natural products that inhibit the catalytic function of human tyrosyl-DNA phosphodiesterase (TDP1). SLAS Discov 22:1093–1105. doi: 10.1177/2472555217717200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Beutler JA, Alvarado-Lindner AB and McCloud TG 1996. Further studies on phorbol ester bioactivity in the Euphorbiaceae. Ann. Missouri Bot. Garden 83:530–533. doi: 10.2307/2399992. [DOI] [Google Scholar]
  17. Beutler JA, Alvarado AB, McCloud TG and Cragg GM 1989. Distribution of phorbol ester bioactivity in the Euphorbiaceae. Phytother. Res 3:188–192. doi: 10.1002/ptr.2650030507. [DOI] [Google Scholar]
  18. Beutler JA, Alvarado AB, Schaufelberger DE, Andrews P and McCloud TG 1990. Dereplication of phorbol bioactives: Lyngbya majuscula and Croton cuneatus. J. Nat. Prod 53:867–874. doi: 10.1021/np50070a014. [DOI] [PubMed] [Google Scholar]
  19. Beutler JA, Karbon EW, Brubaker AN, Malik R, Curtis DR and Enna SJ 1985. Securinine alkaloids: A new class of GABA receptor antagonist. Brain Res. 330:135–140. doi: 10.1016/0006-8993(85)90014-9. [DOI] [PubMed] [Google Scholar]
  20. Beutler JA, Kashman Y, Tischler M, Cardellina JH II, Gray GN, Currens M, Wall ME, Wani MC and Boyd MR 1995. A reinvestigation of Maprounea triterpenes. J. Nat. Prod 58:1039–1046. doi: 10.1021/np50121a008. [DOI] [PubMed] [Google Scholar]
  21. Beutler JA, McKee TC, Fuller RW, Tischler M, Cardellina JH II, Snader KM, McCloud TG and Boyd MR 1993. Frequent occurrence of HIV-inhibitory sulphated polysaccharides in marine invertebrates. Antiviral Chem. Chemother 4:167–172. doi: 10.1177/095632029300400306. [DOI] [Google Scholar]
  22. Bindseil KU, Jakupovic J, Wolf D, Lavayre J, Leboul J and van der PD 2001. Pure compound libraries; a new perspective for natural product based drug discovery. Drug Discov. Today 6:840–847. doi: 10.1016/S1359-6446(01)01856-6. [DOI] [PubMed] [Google Scholar]
  23. Bladt S and Wagner H 2007. From the Zulu medicine to the European phytomedicine Umckaloabo. Phytomedicine 14 Suppl 6:2–4. doi: 10.1016/j.phymed.2006.11.030. [DOI] [PubMed] [Google Scholar]
  24. Blees JS, Schmid T, Thomas CL, Baker AR, Benson L, Evans JR, Goncharova EI, Colburn NH, McMahon JB and Henrich CJ 2010. Development of a high-throughput cell-based reporter assay to identify stabilizers of tumor suppressor Pdcd4. J.Biomol.Screen 15:21–29. doi: 10.1177/1087057109351028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Booth NL, Sayers TJ, Brooks AD, Thomas CF, Jacobsen K, Goncharova EI, McMahon JB, Henrich CJ and Henrich CJ 2009. A cell-based high-throughput screen to identify synergistic TRAIL sensitizers. Cancer Immunol.Immunother 58:1229–1244. doi: 10.1007/s00262-008-0637-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Borchardt JK 1996. A short history of quinine. Drug News & Persp. 9:116–120. doi: 0. [Google Scholar]
  27. Boyd MR, Farina C, Belfiore P, Gagliardi S, Kim JW, Hayakawa Y, Beutler JA, McKee TC, Bowman BJ and Bowman EJ 2001. Discovery of a novel antitumor benzolactone enamide class that selectively inhibits mammalian vacuolar-type (H+)-ATPases. J. Pharmacol. Exp. Ther 297:114–120. doi: 0. [PubMed] [Google Scholar]
  28. Brehm L, Hjeds H and Krogsgaard-Larsen P 1972. The structure of muscimol, a GABA analogue of restricted conformation. Acta Chem. Scand 26:1298–1299. doi: 0. [DOI] [PubMed] [Google Scholar]
  29. Brkljaca R, Schneider B, Hidalgo W, Otalvaro F, Ospina F, Lee S, Hoshino M, Fujita M and Urban S 2017. Application of the crystalline sponge method to revise the structure of the phenalenone fuliginone. Molecules 22. doi: 10.3390/molecules22020211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Brown A and Clardy J 2018. Tiny crystals have big potential for determining structures of small molecules. Nature 564:348–349. doi: 10.1038/d41586-018-07756-5. [DOI] [PubMed] [Google Scholar]
  31. Bucar F, Wube A and Schmid M 2013. Natural product isolation--how to get from biological material to pure compounds. Nat Prod Rep 30:525–545. doi: 10.1039/c3np20106f. [DOI] [PubMed] [Google Scholar]
  32. Buckingham J and Thompson S 1997. The Dictionary of Natural Products and other information sources for natural products scientists Phytochemical Diversity - A Source of New Industrial Products. (Wrigley S, Hayes M, Thomas R & Chrystal E, eds). pp. 53–67. Royal Society of Chemistry, London. [Google Scholar]
  33. Buedenbender L, Habener LJ, Grkovic T, Kurtboke DI, Duffy S, Avery VM and Carroll AR 2018. HSQC-TOCSY fingerprinting for prioritization of polyketide- and peptide-producing microbial isolates. J. Nat. Prod 81:957–965. doi: 10.1021/acs.jnatprod.7b01063. [DOI] [PubMed] [Google Scholar]
  34. Buenz EJ, Verpoorte R and Bauer BA 2018. The ethnopharmacologic contribution to bioprospecting natural products. Annu. Rev. Pharmacol. Toxicol 58:509–530. doi: 10.1146/annurev-pharmtox-010617-052703. [DOI] [PubMed] [Google Scholar]
  35. Bugni TS, Harper MK, McCulloch MW, Reppart J and Ireland CM 2008. Fractionated marine invertebrate extract libraries for drug discovery. Molecules 13:1372–1383. doi: 10.3390/molecules13061372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Butler LG 1992. Antinutritional effects of condensed and hydrolyzable tannins. 2nd North American Tannin Conference on Plant Polyphenols: Biogenesis, Chemical Properties, and Significance, Houghton, MI, USA, Plenum. [Google Scholar]
  37. Butler MS, Robertson AA and Cooper MA 2014. Natural product and natural product derived drugs in clinical trials. Nat. Prod. Rep 31:1612–1661. doi: 10.1039/c4np00064a. [DOI] [PubMed] [Google Scholar]
  38. Cardellina JH II, Munro MHG, Fuller RW, Manfredi KP, McKee TC, Tischler M, Bokesch HR, Gustafson KR, Beutler JA and Boyd MR 1993. A chemical screening strategy for the dereplication and prioritization of HIV-inhibitory aqueous natural products extracts. J. Nat. Prod 56:1123–1129. doi: 10.1021/np50097a016. [DOI] [PubMed] [Google Scholar]
  39. Carroll AR, Copp BR, Davis RA, Keyzers RA and Prinsep MR 2019. Marine natural products. Nat. Prod. Rep 36:122–173. doi: 10.1039/c8np00092a. [DOI] [PubMed] [Google Scholar]
  40. Caty SN, Alvarez-Buylla A, Byrd GD, Vidoudez C, Roland AB, Tapia EE, Budnik B, Trauger SA, Coloma LA and O’Connell LA 2019. Molecular physiology of chemical defenses in a poison frog. J Exp Biol. doi: 10.1242/jeb.204149. [DOI] [PubMed] [Google Scholar]
  41. Chain E, Florey HW, Gardner AD, Heatley NG, Jennings MA, Orr-Ewing J and Sanders AG 1940. Penicillin as a chemotherapeutic agent. Lancet 236:226–228. doi: 0. [Google Scholar]
  42. Chordia P and MacArthur RD 2013. Crofelemer, a novel agent for treatment of non-infectious diarrhea in HIV-infected persons. Expert Rev. Gastroenterol. Hepatol 7:591–600. doi: 10.1586/17474124.2013.832493. [DOI] [PubMed] [Google Scholar]
  43. Cianfrocca M 2008. Application of epothilones in breast cancer therapy. Curr Opin Oncol 20:634–638. doi: 10.1097/CCO.0b013e32831270b0. [DOI] [PubMed] [Google Scholar]
  44. ClinicalTrials.gov. A phase III trial of Marizomib in patients with newly diagnosed glioblastoma. https://clinicaltrials.gov/ct2/show/NCT03345095. Accessed 6/25/2019.
  45. Corley DG and Durley RC 1994. Strategies for database dereplication of natural products. J. Nat. Prod 57:1484–1490. doi: 10.1021/np50113a002. [DOI] [Google Scholar]
  46. Covington BC, McLean JA and Bachmann BO 2017. Comparative mass spectrometry-based metabolomics strategies for the investigation of microbial secondary metabolites. Nat. Prod. Rep 34:6–24. doi: 10.1039/c6np00048g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Cragg GM, Katz F, Newman DJ and Rosenthal J 2012. The impact of the United Nations Convention on Biological Diversity on natural products research. Nat. Prod. Rep 29:1407–1423. doi: 10.1039/c2np20091k. [DOI] [PubMed] [Google Scholar]
  48. Cragg GM and Newman DJ 2005. International collaboration in drug discovery and development from natural sources. Pure Appl. Chem 77:1923–1942. doi: 10.1351/pac200577111923. [DOI] [Google Scholar]
  49. Cravens A, Payne J and Smolke CD 2019. Synthetic biology strategies for microbial biosynthesis of plant natural products. Nat. Commun 10:2142. doi: 10.1038/s41467-019-09848-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Crits-Christoph A, Diamond S, Butterfield CN, Thomas BC and Banfield JF 2018. Novel soil bacteria possess diverse genes for secondary metabolite biosynthesis. Nature 558:440–444. doi: 10.1038/s41586-018-0207-y. [DOI] [PubMed] [Google Scholar]
  51. da Cunha BR, Fonseca LP and Calado CRC 2019. Antibiotic discovery: Where have we come from, where do we go? Antibiotics 8:45. doi: 0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Dabydeen DA, Burnett JC, Bai R, Verdier-Pinard P, Hickford SJH, Pettit GR, Blunt JW, Munro MHG, Gussio R and Hamel E 2006. Comparison of the activities of the truncated halichondrin B analog NSC 707389 (E7389) with those of the parent compound and a proposed binding site on tubulin. Mol. Pharmacol 70:1866–1875. doi: 10.1124/mol.106.026641. [DOI] [PubMed] [Google Scholar]
  53. Daly JW, Spande TF and Garraffo HM 2005. Alkaloids from amphibian skin: A tabulation of over eight-hundred compounds. J. Nat. Prod 68:1556–1575. doi: 10.1021/np0580560. [DOI] [PubMed] [Google Scholar]
  54. Davies-Coleman MT, Faulkner DJ, Dubowchik GM, Roth GP, Polson C and Fairchild C 1993. A New EGF-active polymeric pyridinium alkaloid from the sponge Callyspongia fibrosa. J. Org. Chem 58:5925–5930. doi: 10.1021/jo00074a017. [DOI] [Google Scholar]
  55. Deocaris CC, Widodo N, Wadhwa R and Kaul SC 2008. Merger of ayurveda and tissue culture-based functional genomics: Inspirations from systems biology. J. Transl. Med 6:14. doi: 10.1186/1479-5876-6-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Dinan L and Lafont R 2006. Effects and applications of arthropod steroid hormones (ecdysteroids) in mammals. J. Endocrinol 191:1–8. doi: 10.1677/joe.1.06900. [DOI] [PubMed] [Google Scholar]
  57. Donadio S, Staver MJ, McAlpine JB, Swanson SJ and Katz L 1991. Modular organization of genes required for complex polyketide biosynthesis. Science 252:675–679. doi: 10.1126/science.2024119. [DOI] [PubMed] [Google Scholar]
  58. Dorn BR 2013. Mayo v. Prometheus: A year later. ACS Med. Chem. Lett 4:572–573. doi: 10.1021/ml400230u. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. El-Elimat T, Figueroa M, Raja HA, Graf TN, Swanson SM, Falkinham JO III, Wani MC, Pearce CJ and Oberlies NH 2015. Biosynthetically distinct cytotoxic polyketides from Setophoma terrestris. Eur. J. Org. Chem 2015:109–121. doi: 10.1002/ejoc.201402984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Eldridge GR, Vervoort HC, Lee CM, Cremin PA, Williams CT, Hart SM, Goering MG, O’Neil-Johnson M and Zeng L 2002. High-throughput method for the production and analysis of large natural product libraries for drug discovery. Anal. Chem 74:3963–3971. doi: 10.1021/ac025534s. [DOI] [PubMed] [Google Scholar]
  61. Epstein S and López-Garcia P 2008. “Missing” protists: A molecular prospective. Biodivers. Conserv 17:261–276. doi: 10.1007/s10531-007-9250-y. [DOI] [Google Scholar]
  62. Feng X and Siegel MM 2007. FTICR-MS applications for the structure determination of natural products. Anal. Bioanal. Chem 389:1341–1363. doi: 10.1007/s00216-007-1468-8. [DOI] [PubMed] [Google Scholar]
  63. Florez LV, Biedermann PH, Engl T and Kaltenpoth M 2015. Defensive symbioses of animals with prokaryotic and eukaryotic microorganisms. Nat. Prod. Rep 32:904–936. doi: 10.1039/c5np00010f. [DOI] [PubMed] [Google Scholar]
  64. Gaedcke F 1855. Ueber das erythroxylin, dargestellt aus den blättern des in Südamerika cultivirten strauches Erythroxylon coca Lam. Archiv der Pharmazie 132:141–150. doi: 10.1002/ardp.18551320208. [DOI] [Google Scholar]
  65. Garcia A, Drown BS and Hergenrother PJ 2016. Access to a structurally complex compound collection via ring distortion of the alkaloid sinomenine. Org Lett 18:4852–4855. doi: 10.1021/acs.orglett.6b02333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Gates M and Tschudi G 1956. The synthesis of morphine. J. Am. Chem. Soc 78:1380–1393. doi: 10.1021/ja01588a033. [DOI] [Google Scholar]
  67. Gay GM, Inaba DS, Sheppard CW, Newmeyer JA and Rappolt RT 1975. Cocaine: History, epidemiology, human pharmacology, and treatment. A perspective on a new debut for an old girl. Clin. Toxicol 8:149–178. doi: 10.3109/15563657508988061. [DOI] [PubMed] [Google Scholar]
  68. Golik J, Dubay G, Groenwald G, Kawaguchi H, Konishi M, Krishnan B, Ohkuma H, Saitoh K and Doyle TW 1987. Esperamicins, a novel class of potent antitumor antibiotics. 3. Structures of esperamicins A1, A2 and A1b. J. Am. Chem. Soc 109:3464. doi: 10.1021/ja00245a049. [DOI] [PubMed] [Google Scholar]
  69. Gordon J 2014. The impact of Myriad and Mayo: Will advancements in the biological sciences be spurred or disincentivized? (Or was biotech patenting not complicated enough?). Cold Spring Harb. Perspect. Med 5:a020917. doi: 10.1101/cshperspect.a020917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Gruene T, Wennmacher JTC, Zaubitzer C, Holstein JJ, Heidler J, Fecteau-Lefebvre A, De Carlo S, Muller E, Goldie KN, Regeni I, Li T, Santiso-Quinones G, Steinfeld G, Handschin S, van Genderen E, van Bokhoven JA, Clever GH and Pantelic R 2018. Rapid structure determination of microcrystalline molecular compounds using electron diffraction. Angew. Chem. Int. Ed. Engl 57:16313–16317. doi: 10.1002/anie.201811318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Gulland JM and Robinson R 1923. The morphine group. Part I. A discussion of the constitutional problem. J. Chem. Soc:980–998. doi: 10.1039/ct9232300980. [DOI] [Google Scholar]
  72. Gullo VP and Hughes DE 2005. Exploiting new approaches for natural product drug discovery in the biotechnology industry. Drug Discovery Today 2:281–286. doi: 10.1016/j.ddtec.2005.08.002. [DOI] [PubMed] [Google Scholar]
  73. Hartwell JL 1967. Plants used against cancer. A survey. Lloydia 30:379–436. doi: 0. [PubMed] [Google Scholar]
  74. Harvey CJB, Tang M, Schlecht U, Horecka J, Fischer CR, Lin HC, Li J, Naughton B, Cherry J, Miranda M, Li YF, Chu AM, Hennessy JR, Vandova GA, Inglis D, Aiyar RS, Steinmetz LM, Davis RW, Medema MH, Sattely E, Khosla C, St Onge RP, Tang Y and Hillenmeyer ME 2018. HEx: A heterologous expression platform for the discovery of fungal natural products. Sci Adv 4:eaar5459. doi: 10.1126/sciadv.aar5459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hemmilä I and Webb S 1997. Time-resolved fluorometry: An overview of the labels and core technologies for drug screening applications. Drug Discovery Today 2:373–381. doi: 10.1016/S1359-6446(97)01080-5. [DOI] [Google Scholar]
  76. Henrich CJ, Bokesch HR, Dean M, Bates SE, Robey RW, Goncharova EI, Wilson JA and McMahon JB 2006. A high-throughput cell-based assay for inhibitors of ABCG2 activity. J.Biomol.Screen 11:176–183. doi: 10.1177/1087057105284576. [DOI] [PubMed] [Google Scholar]
  77. Hildebrand M, Waggoner LE, Liu H, Sudek S, Allen S, Anderson C, Sherman DH and Haygood M 2004. bryA: An unusual modular polyketide synthase gene from the uncultivated bacterial symbiont of the marine bryozoan Bugula neritina. Chem. Biol 11:1543–1552. doi: 10.1016/j.chembiol.2004.08.018. [DOI] [PubMed] [Google Scholar]
  78. Holodniy M, Koch J, Mistal M, Maimares Schmidt J, Khandwala A, Pennington JE and Porter SB 1999. A double-blind, randomized, placebo-controlled phase II study to assess the safety and efficacy of orally administered SP-303 for the symptomatic treatment of diarrhea in patients with AIDS. Am. J. Gastroenterol 94:3267–3273. doi: 10.1016/S0002-9270(99)00594–8. [DOI] [PubMed] [Google Scholar]
  79. Hoshino M, Khutia A, Xing H, Inokuma Y and Fujita M 2016. The crystalline sponge method updated. Int. U. Crystallogr. J 3:139–151. doi: 10.1107/S2052252515024379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hostettmann K, Hostettmann M and Marston A 1998. Preparative Chromatography Techniques Applications in Natural Products Isolation. Berlin, Springer. [Google Scholar]
  81. Hostettmann K and Marston A 1995. Saponins. Cambridge, UK, Cambridge University Press. [Google Scholar]
  82. Huyck TK, Gradishar W, Manuguid F and Kirkpatrick P 2011. Eribulin mesylate. Nat. Rev. Drug Discov 10:173–174. doi: 10.1038/nrd3389. [DOI] [PubMed] [Google Scholar]
  83. Inokuma Y, Yoshioka S, Ariyoshi J, Arai T, Hitora Y, Takada K, Matsunaga S, Rissanen K and Fujita M 2013. X-ray analysis on the nanogram to microgram scale using porous complexes. Nature 495:461–466. doi: 10.1038/nature11990. [DOI] [PubMed] [Google Scholar]
  84. Johnston GAR, Beart PM, Curtis DR, Game CJA, McCulloch RM and MacLachlan RM 1972. Bicuculline methochloride as a GABA antagonist. Nature New Biol. 240:219–220. doi: 10.1038/newbio240219a0. [DOI] [PubMed] [Google Scholar]
  85. Jones CG, Martynowycz MW, Hattne J, Fulton T, Stoltz BM, Rodriguez JA, Nelson HM and Gonen T 2018. The CryoEM method MicroED as a powerful tool for small molecule structure determination. ACS Central Sci. 4:1587–1592. doi: 10.1021/acscentsci.8b00760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Joppa LN, Visconti P, Jenkins CN and Pimm SL 2013. Achieving the convention on biological diversity’s goals for plant conservation. Science 341:1100–1103. doi: 341/6150/1100 [pii]; 10.1126/science.1241706 [doi]. [DOI] [PubMed] [Google Scholar]
  87. Kai K, Sogame M, Sakurai F, Nasu N and Fujita M 2018. Collimonins A-D, unstable polyynes with antifungal or pigmentation activities from the fungus-feeding bacterium Collimonas fungivorans Ter331. Org. Lett 20:3536–3540. doi: 10.1021/acs.orglett.8b01311. [DOI] [PubMed] [Google Scholar]
  88. Katz L and Baltz RH 2016. Natural product discovery: past, present, and future. J. Ind. Microbiol. Biotechnol 43:155–176. doi: 10.1007/s10295-015-1723-5. [DOI] [PubMed] [Google Scholar]
  89. Kearney SE, Zahoranszky-Kohalmi G, Brimacombe KR, Henderson MJ, Lynch C, Zhao T, Wan KK, Itkin Z, Dillon C, Shen M, Cheff DM, Lee TD, Bougie D, Cheng K, Coussens NP, Dorjsuren D, Eastman RT, Huang R, Iannotti MJ, Karavadhi S, Klumpp-Thomas C, Roth JS, Sakamuru S, Sun W, Titus SA, Yasgar A, Zhang YQ, Zhao J, Andrade RB, Brown MK, Burns NZ, Cha JK, Mevers EE, Clardy J, Clement JA, Crooks PA, Cuny GD, Ganor J, Moreno J, Morrill LA, Picazo E, Susick RB, Garg NK, Goess BC, Grossman RB, Hughes CC, Johnston JN, Joullie MM, Kinghorn AD, Kingston DGI, Krische MJ, Kwon O, Maimone TJ, Majumdar S, Maloney KN, Mohamed E, Murphy BT, Nagorny P, Olson DE, Overman LE, Brown LE, Snyder JK, Porco JA Jr., Rivas F, Ross SA, Sarpong R, Sharma I, Shaw JT, Xu Z, Shen B, Shi W, Stephenson CRJ, Verano AL, Tan DS, Tang Y, Taylor RE, Thomson RJ, Vosburg DA, Wu J, Wuest WM, Zakarian A, Zhang Y, Ren T, Zuo Z, Inglese J, Michael S, Simeonov A, Zheng W, Shinn P, Jadhav A, Boxer MB, Hall MD, Xia M, Guha R and Rohde JM 2018. Canvass: A crowd-sourced, natural-product screening library for exploring biological space. ACS Cent. Sci 4:1727–1741. doi: 10.1021/acscentsci.8b00747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Keskin O, Bahar I, Jernigan RL, Myers TG, Beutler JA, Shoemaker RH, Sausville EA and Covell DG 2000. Characterization of anticancer agents by their growth-inhibitory activity and relationships to mechanism of action and structure. Anticancer Drug Des. 15:79–98. doi: 0. [PubMed] [Google Scholar]
  91. Khanbabaee K and Ree T. v. 2001. Tannins: Classification and definition. Nat. Prod. Rep 18:641–649. doi: 10.1039/b101061l. [DOI] [PubMed] [Google Scholar]
  92. Kupče Ē and Claridge TDW 2017. NOAH: NMR supersequences for small molecule analysis and structure elucidation. Angew. Chem. Int. Ed. Engl 56:11779–11783. doi: 10.1002/anie.201705506. [DOI] [PubMed] [Google Scholar]
  93. Lang G, Mayhudin NA, Mitova MI, Sun L, van der SS, Blunt JW, Cole AL, Ellis G, Laatsch H and Munro MH 2008. Evolving trends in the dereplication of natural product extracts: new methodology for rapid, small-scale investigation of natural product extracts. J. Nat. Prod 71:1595–1599. doi: 10.1021/np8002222. [DOI] [PubMed] [Google Scholar]
  94. Lee MK 2015. July 2015 update on subject matter eligibility. Fed. Reg 80:45429–45429. doi: 0. [Google Scholar]
  95. Lee MK 2016. May 2016 subject matter eligibility update. Fed. Reg 81:27381–27382. doi: 0. [Google Scholar]
  96. Lee S, Hoshino M, Fujita M and Urban S 2017. Cycloelatanene A and B: Absolute configuration determination and structural revision by the crystalline sponge method. Chem. Sci 8:1547–1550. doi: 10.1039/c6sc04288k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Lerchen HG 2002. Milestones in camptothecin research. Drugs Fut. 27:869–878. doi: 10.1358/dof.2002.027.09.694498. [DOI] [Google Scholar]
  98. Li K, Yang DS, Gu XF and Di B 2019. Absolute configuration determination of asarinin by synchrotron radiation with crystalline sponge method. Fitoterapia 134:135–140. doi: 10.1016/j.fitote.2019.02.010. [DOI] [PubMed] [Google Scholar]
  99. Li Z, Nakashige M and Chain WJ 2011. A brief synthesis of (−)-englerin A. J. Am. Chem. Soc 133:6553–6556. doi: 10.1021/ja201921j. [DOI] [PubMed] [Google Scholar]
  100. Lidert Z, Wing K, Polonsky J, Imakura Y, Masayoshi Okano, Tani S, Lin YM, Kiyokawa H and Lee KH 1987. Insect antifeedant and growth inhibitory activity of forty-six quassinoids on two species of agricultural pests. J. Nat. Prod 50:442–448. doi: 10.1021/np50051a016. [DOI] [Google Scholar]
  101. Lipinski CA, Lombardo F, Dominy BW and Feeney PJ 1997. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev 23:3–25. doi: 10.1016/S0169-409X(96)00423-1. [DOI] [PubMed] [Google Scholar]
  102. Liu Y, Cohen RD, Gustafson KR, Martin GE and Williamson RT 2018. Enhanced measurement of residual chemical shift anisotropy for small molecule structure elucidation. Chem Commun 54:4254–4257. doi: 10.1039/c8cc00552d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Lockemann G 1951. Friedrich Wilhelm Serturner, the discoverer of morphine. J. Chem. Ed 28:279. doi: 0. [Google Scholar]
  104. Lopez JV, Kamel B, Medina M, Collins T and Baums IB 2019. Multiple facets of marine invertebrate conservation genomics. Annu Rev Anim Biosci 7:473–497. doi: 10.1146/annurev-animal-020518-115034. [DOI] [PubMed] [Google Scholar]
  105. Luo X, Reiter MA, d’Espaux L, Wong J, Denby CM, Lechner A, Zhang Y, Grzybowski AT, Harth S, Lin W, Lee H, Yu C, Shin J, Deng K, Benites VT, Wang G, Baidoo EEK, Chen Y, Dev I, Petzold CJ and Keasling JD 2019. Complete biosynthesis of cannabinoids and their unnatural analogues in yeast. Nature 567:123–126. doi: 10.1038/s41586-019-0978-9. [DOI] [PubMed] [Google Scholar]
  106. Matsuda Y, Mitsuhashi T, Lee S, Hoshino M, Mori T, Okada M, Zhang H, Hayashi F, Fujita M and Abe I 2016. Astellifadiene: Structure determination by NMR spectroscopy and crystalline sponge method, and elucidation of its biosynthesis. Angew. Chem. Int. Ed. Engl 55:5785–5788. doi: 10.1002/anie.201601448. [DOI] [PubMed] [Google Scholar]
  107. McAlpine JB, Chen SN, Kutateladze A, MacMillan JB, Appendino G, Barison A, Beniddir MA, Biavatti MW, Bluml S, Boufridi A, Butler MS, Capon RJ, Choi YH, Coppage D, Crews P, Crimmins MT, Csete M, Dewapriya P, Egan JM, Garson MJ, Genta-Jouve G, Gerwick WH, Gross H, Harper MK, Hermanto P, Hook JM, Hunter L, Jeannerat D, Ji NY, Johnson TA, Kingston DGI, Koshino H, Lee HW, Lewin G, Li J, Linington RG, Liu M, McPhail KL, Molinski TF, Moore BS, Nam JW, Neupane RP, Niemitz M, Nuzillard JM, Oberlies NH, Ocampos FMM, Pan G, Quinn RJ, Reddy DS, Renault JH, Rivera-Chavez J, Robien W, Saunders CM, Schmidt TJ, Seger C, Shen B, Steinbeck C, Stuppner H, Sturm S, Taglialatela-Scafati O, Tantillo DJ, Verpoorte R, Wang BG, Williams CM, Williams PG, Wist J, Yue JM, Zhang C, Xu Z, Simmler C, Lankin DC, Bisson J and Pauli GF 2019. The value of universally available raw NMR data for transparency, reproducibility, and integrity in natural product research. Nat. Prod. Rep 36:35–107. doi: 10.1039/c7np00064b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. McCloud TG 2010. High throughput extraction of plant, marine and fungal specimens for preservation of biologically active molecules. Molecules 15:4526–4563. doi: 10.3390/molecules15074526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. McKee TC, Covington CD, Fuller RW, Bokesch HR, Young S, Cardellina JH II, Kadushin MR, Soejarto DD, Stevens PF, Cragg GM and Boyd MR 1998a. Pyranocoumarins from tropical species of the genus Calophyllum: A chemotaxonomic study of extracts in the National Cancer Institute collection. J. Nat. Prod 61:1252–1256. doi: 10.1021/np980140a. [DOI] [PubMed] [Google Scholar]
  110. McKee TC, Galinis DL, Pannell LK, Cardellina JH II, Laakso J, Ireland CM, Murray L, Capon RJ and Boyd MR 1998b. The lobatamides, novel cytotoxic macrolides from southwestern Pacific tunicates. J. Org. Chem 63:7805–7810. doi: 10.1021/JO980939R. [DOI] [Google Scholar]
  111. Mendola D 2003. Aquaculture of three phyla of marine invertebrates to yield bioactive metabolites: Process developments and economics. Biomol. Eng 20:441–458. doi: 10.1016/S1389-0344(03)00075-3. [DOI] [PubMed] [Google Scholar]
  112. Milanowski DJ, Oku N, Cartner LK, Bokesch HR, Williamson RT, Sauri J, Liu Y, Blinov KA, Ding Y, Li XC, Ferreira D, Walker LA, Khan S, Davies-Coleman MT, Kelley JA, McMahon JB, Martin GE and Gustafson KR 2018. Unequivocal determination of caulamidines A and B: application and validation of new tools in the structure elucidation tool box. Chem. Sci 9:307–314. doi: 10.1039/c7sc01996c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Miller IJ, Vanee N, Fong SS, Lim-Fong GE and Kwan JC 2016. Lack of overt genome reduction in the bryostatin-producing bryozoan symbiont “Candidatus Endobugula sertula”. Appl. Environ. Microbiol 82:6573–6583. doi: 10.1128/AEM.01800-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Molawai K, Delpont N and Echavarren AM 2010. Enantioselective synthesis of (−)-englerins A and B. Angew. Chem. Int. Ed. Engl 49:3517–3519. doi: 10.1002/anie.201000890. [DOI] [PubMed] [Google Scholar]
  115. Møller BL and Ratcliffe RG 2014. Editorial overview: Synthetic plant biology: The roots of a bio-based society. Curr. Opin. Biotechnol 26:ix–xvi. doi: 10.1016/j.copbio.2014.02.016. [DOI] [PubMed] [Google Scholar]
  116. Moriya T, Saito K, Kurita H, Matsumoto K, Otake T, Mori H, Morimoto M, Ueba N and Kunita N 1993. A new candidate for an anti-HIV-1 agent: modified cyclodextrin sulfate (mCDS71). J. Med. Chem 36:1674–1677. doi: 10.1021/jm00063a018. [DOI] [PubMed] [Google Scholar]
  117. Mothes K 1969. Die alkaloide im stoffwechsel der pflanze. Experientia 25:225–239. doi: 0. [DOI] [PubMed] [Google Scholar]
  118. Mullin R Merck cuts sales jobs and halts natural products research. Chem. & Eng. News, May 12, 2008, p.21. [Google Scholar]
  119. Munro MH, Blunt JW, Dumdei EJ, Hickford SJ, Lill RE, Li S, Battershill CN and Duckworth AR 1999. The discovery and development of marine compounds with pharmaceutical potential. J. Biotechnol 70:15–25. doi: 10.1016/S0168-1656(99)00052-8. [DOI] [PubMed] [Google Scholar]
  120. AMP v. Myriad. United States Supreme Court. Association for Molecular Pathology et al. v. Myriad Genetics, Inc, et al. Docket no. 12–398, Decided June 13, 2013. [Google Scholar]
  121. Nannenga BL and Gonen T 2019. The cryo-EM method microcrystal electron diffraction (MicroED). Nat Methods 16:369–379. doi: 10.1038/s41592-019-0395-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Nass R, Merchant KM and Ryan T 2008. Caenorhabditis elegans in Parkinson’s Disease drug discovery: Addressing an unmet medical need. Molecular Interventions 8:284–293. doi: 10.1124/mi.8.6.6. [DOI] [PubMed] [Google Scholar]
  123. Newman DJ and Cragg GM 2016. Natural products as sources of new drugs from 1981 to 2014. J Nat Prod 79:629–661. doi: 10.1021/acs.jnatprod.5b01055. [DOI] [PubMed] [Google Scholar]
  124. Nishizuka Y 1984. The role of protein kinase C in cell surface signal transduction and tumour promotion. Nature 308:693–698. doi: 10.1038/308693a0. [DOI] [PubMed] [Google Scholar]
  125. Nothias LF, Nothias-Esposito M, da Silva R, Wang M, Protsyuk I, Zhang Z, Sarvepalli A, Leyssen P, Touboul D, Costa J, Paolini J, Alexandrov T, Litaudon M and Dorrestein PC 2018. Bioactivity-based molecular networking for the discovery of drug leads in natural product bioassay-guided fractionation. J Nat Prod 81:758–767. doi: 10.1021/acs.jnatprod.7b00737. [DOI] [PubMed] [Google Scholar]
  126. O’ Connor CJ, Beckmann HS and Spring DR 2012. Diversity-oriented synthesis: Producing chemical tools for dissecting biology. Chem.Soc.Rev 41:4444–4456. doi: 10.1039/c2cs35023h. [DOI] [PubMed] [Google Scholar]
  127. Olivon F, Allard PM, Koval A, Righi D, Genta-Jouve G, Neyts J, Apel C, Pannecouque C, Nothias LF, Cachet X, Marcourt L, Roussi F, Katanaev VL, Touboul D, Wolfender JL and Litaudon M 2017. Bioactive natural products prioritization using massive multi-informational molecular networks. ACS chemical biology 12:2644–2651. doi: 10.1021/acschembio.7b00413. [DOI] [PubMed] [Google Scholar]
  128. Olivon F, Remy S, Grelier G, Apel C, Eydoux C, Guillemot JC, Neyts J, Delang L, Touboul D, Roussi F and Litaudon M 2019. Antiviral compounds from Codiaeum peltatum targeted by a multi-informative molecular networks approach. J Nat Prod 82:330–340. doi: 10.1021/acs.jnatprod.8b00800. [DOI] [PubMed] [Google Scholar]
  129. Paddon CJ, Westfall PJ, Pitera DJ, Benjamin K, Fisher K, McPhee D, Leavell MD, Tai A, Main A, Eng D, Polichuk DR, Teoh KH, Reed DW, Treynor T, Lenihan J, Fleck M, Bajad S, Dang G, Dengrove D, Diola D, Dorin G, Ellens KW, Fickes S, Galazzo J, Gaucher SP, Geistlinger T, Henry R, Hepp M, Horning T, Iqbal T, Jiang H, Kizer L, Lieu B, Melis D, Moss N, Regentin R, Secrest S, Tsuruta H, Vazquez R, Westblade LF, Xu L, Yu M, Zhang Y, Zhao L, Lievense J, Covello PS, Keasling JD, Reiling KK, Renninger NS and Newman JD 2013. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature 496:528–532. doi: 10.1038/nature12051. [DOI] [PubMed] [Google Scholar]
  130. Paull KD, Lin CM, Malspeis L and Hamel E 1992. Identification of novel antimitotic agents acting at the tubulin level by computer-assisted evaluation of differential cytotoxicity data. Cancer Res. 52:3892–3900. doi: 0. [PubMed] [Google Scholar]
  131. Paull KD, Shoemaker RH, Hodes L, Monks A, Scudiero DA, Rubinstein L, Plowman J and Boyd MR 1989. Display and analysis of patterns of differential activity of drugs against human tumor cell lines: Development of mean graph and COMPARE algorithm. J. Natl. Cancer Inst 81:1088–1092. doi: 10.1093/jnci/81.14.1088. [DOI] [PubMed] [Google Scholar]
  132. Pelletier PJ and Caventou JB 1820. Suite: Des recherches chimiques sur les quinquinas. Ann. Chim. Phys 15:337–365. doi: 0. [Google Scholar]
  133. Pettit GR, Herald CL, Boyd MR, Leet JE, Dufresne C, Doubek DL, Schmidt JM, Cerny RL, Hooper JN and Rutzler KC 1991. Isolation and structure of the cell growth inhibitory constituents from the Pacific marine sponge Axinella sp. J. Med. Chem 34:3339–3340. doi: 10.1021/jm00115a027. [DOI] [PubMed] [Google Scholar]
  134. Pettit GR, Inoue M, Kamano Y, Herald DL, Arm C, Dufresne C, Christie ND, Schmidt JM, Doubek DL and Krupa TS 1988. Isolation and structure of the powerful cell growth inhibitor cephalostatin 1. J.Am.Chem.Soc 110:2006–2007. doi: [Google Scholar]
  135. Piel J 2002. A polyketide synthase-peptide synthetase gene cluster from an uncultured bacterial symbiont of Paederus beetles. Proc. Natl. Acad. Sci. U. S. A 99:14002–14007. doi: 10.1073/pnas.222481399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Mayo v. Prometheus. United States Supreme Court. Mayo Collaborative Services d/b/a Mayo Medical Laboratories, et al. v. Prometheus Laboratories, Inc; Docket no. 10–1150, Decided March 20, 2012. [Google Scholar]
  137. Puglisi MP, Sneed JM, Ritson-Williams R and Young R 2019. Marine chemical ecology in benthic environments. Nat. Prod. Rep 36:410–429. doi: 10.1039/c8np00061a. [DOI] [PubMed] [Google Scholar]
  138. Rabe P 1908. Information on the china alcaloide. VIII. Announcement: The constitution of cinchonines. Ber. Deutsch. Chem. Ges 41:62–70. doi: 0. [Google Scholar]
  139. Raffauf RF 1996. Plant Alkaloids. A Guide to their Discovery and Distribution. Binghamton, NY, Food Products Press. [Google Scholar]
  140. Ramadhar TR, Zheng SL, Chen YS and Clardy J 2015. Analysis of rapidly synthesized guest-filled porous complexes with synchrotron radiation: practical guidelines for the crystalline sponge method. Acta Crystallogr A Found Adv 71:46–58. doi: 10.1107/S2053273314019573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Ratnayake R, Covell DG, Ransom TT, Gustafson KR and Beutler JA 2009. Englerin A, a selective inhibitor of renal cancer cell growth, from Phyllanthus engleri. Org. Lett 11:57–60. doi: 10.1021/ol802339w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Ravindran S Discovering novel antibiotics. The Scientist, February 1, 2017, p.32100. [Google Scholar]
  143. Reynolds WF and Enriquez RG 2002. Choosing the best pulse sequences, acquisition parameters, postacquisition processing strategies, and probes for natural product structure elucidation by NMR spectroscopy. J. Nat. Prod 65:221–244. doi: 10.1021/np010444o. [DOI] [PubMed] [Google Scholar]
  144. Rompler H, Staubert C, Thor D, Schulz A, Hofreiter M and Schoneberg T 2007. G Protein-coupled time travel: Evolutionary aspects of GPCR research. Molecular Interventions 7:17–25. doi: 10.1124/mi.7.1.5. [DOI] [PubMed] [Google Scholar]
  145. Ruocco KM, Goncharova EI, Young MR, Colburn NH, McMahon JB and Henrich CJ 2007. A high-throughput cell-based assay to identify specific inhibitors of transcription factor AP-1. J Biomol.Screen. 12:133–139. doi: [DOI] [PubMed] [Google Scholar]
  146. Saporito RA, Donnelly MA, Norton RA, Garraffo HM, Spande TF and Daly JW 2007. Oribatid mites as a major dietary source for alkaloids in poison frogs. Proc. Natl. Acad. Sci. U.S. A 104:8885–8890. doi: 10.1073/pnas.0702851104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Sasaki M, Matsumori N, Maruyama T, Nonomura T, Murata M, Tachibana K and Yasumoto T 1996. The complete structure of maitotoxin .1. Configuration of the C1-C14 side chain. Angew Chem Int Ed Engl 35:1672–1675. doi: 10.1002/anie.199616721. [DOI] [Google Scholar]
  148. Schaufelberger DE, Koleck MP, Beutler JA, Vatakis AM, Alvarado AB, Andrews P, Marzo LV, Muschik GM, Roach J, Ross JT, Lebherz WB, Reeves MP, Eberwein RM, Rodgers LL, Testerman RP, Snader KM and Forenza S 1991. The large-scale isolation of bryostatin 1 from Bugula neritina following current Good Manufacturing Practices. J. Nat. Prod 54:1265–1270. doi: 10.1021/np50077a004. [DOI] [PubMed] [Google Scholar]
  149. Schmitz FJ, Hollenbeak KH and Campbell DC 1978. Marine natural products: Halitoxin, toxic complex of several marine sponges of the genus Haliclona. J. Org. Chem 43:3916–3922. doi: 10.1021/jo00414a026. [DOI] [Google Scholar]
  150. Schröder FC, Farndon JR, Attygalle AB, Smedley SR, Eisner T and Meinwald J 1998. Combinatorial chemistry in insects: A library of defensive macrocyclic polyamines. Science 281:428–431. doi: 10.1126/science.281.5375.428. [DOI] [PubMed] [Google Scholar]
  151. Servick K 2015. U.S. patent office reworks unpopular policy. Science 347:113. doi: 10.1126/science.347.6218.113. [DOI] [PubMed] [Google Scholar]
  152. Servick K 2019. Draft bill reignites U.S. debate over patenting human genes. Science 364:1017. doi: 10.1126/science.364.6445.1017. [DOI] [PubMed] [Google Scholar]
  153. Simmons TL, Coates RC, Clark BR, Engene N, Gonzalez D, Esquenazi E, Dorrestein PC and Gerwick WH 2008. Biosynthetic origin of natural products isolated from marine microorganism-invertebrate assemblages. Proc. Natl. Acad. Sci. U. S. A 105:4587–4594. doi: 10.1073/pnas.0709851105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Sittenfeld A, Tamayo G, Nielsen V, Jiménez A, Hurtado P, Chinchilla M, Guerrero O, Mora MA, Rojas M, Blanco R, Alvarado E, Gutiérrez JM and Janzen DH 1999. Costa Rican International Cooperative Biodiversity Group: Using insects and other arthropods in biodiversity prospecting. Pharmaceutical Biol. 37:55–68. doi: 10.1076/1388-0209(200010)37:SUP;1-W;FT055. [DOI] [Google Scholar]
  155. Smith AB and Kim WS 2011. Diversity-oriented synthesis leads to an effective class of bifunctional linchpins uniting anion relay chemistry (ARC) with benzyne reactivity. Proceedings of the National Academy of Sciences 108:6787–6792. doi: 10.1073/pnas.1015265108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Smith SA, Tank DC, Boulanger LA, Bascom-Slack CA, Eisenman K, Kingery D, Babbs B, Fenn K, Greene JS, Hann BD, Keehner J, Kelley-Swift EG, Kembaiyan V, Lee SJ, Li P, Light DY, Lin EH, Ma C, Moore E, Schorn MA, Vekhter D, Nunez PV, Strobel GA, Donoghue MJ and Strobel SA 2008. Bioactive endophytes warrant intensified exploration and conservation. PLoS One 3:e3052. doi: 10.1371/journal.pone.0003052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Soliman SS, Greenwood JS, Bombarely A, Mueller LA, Tsao R, Mosser DD and Raizada MN 2015. An endophyte constructs fungicide-containing extracellular barriers for its host plant. Curr. Biol 25:2570–2576. doi: 10.1016/j.cub.2015.08.027. [DOI] [PubMed] [Google Scholar]
  158. Spjut RW 2005. Relationships between plant folklore and antitumor activity: An historical review. Sida 21:2205–2241. doi: 0. [Google Scholar]
  159. Stevens CL, Taylor KG, Munk ME, Marshall WS, Noll K, Shah GD, Shah LG and Uzu K 1965. Chemistry and structure of mitomycin C. J. Med. Chem 8:1–10. doi: 10.1021/jm00325a001. [DOI] [PubMed] [Google Scholar]
  160. Stierle A, Strobel G and Stierle D 1993. Taxol and taxol production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science 260:214–216. doi: 10.1126/science.8097061. [DOI] [PubMed] [Google Scholar]
  161. Suzumura K, Takahashi I, Matsumoto H, Nagai K, Setiawan B, Rantiatmodjo RM, Suzuki K and Nagano N 1997. Structural elucidation of YM-75518, a novel antifungal antibiotic isolated from Pseudomonas sp. Q38009. Tetrahedron Lett. 38:7573–7576. doi: 10.1016/S0040-4039(97)01785-1. [DOI] [Google Scholar]
  162. Tallmadge EH 2017. Patenting natural products after Myriad. Harvard J Law Technol 30:569–600. doi: 0. [Google Scholar]
  163. Tang P, Mamdani F, Hu X, Liu JO and Yu B 2007. Synthesis of OSW saponin analogs with modified sugar residues and their antiproliferative activities. Bioorg. Med. Chem. Lett 17:1003–1007. doi: 10.1016/j.bmcl.2006.11.032. [DOI] [PubMed] [Google Scholar]
  164. Tang W and Eisenbrand G 1992. Chinese Drugs of Plant Origin: Chemistry, Pharmacology, and Use in Traditional and Modern Medicine. Berlin, Springer-Verlag. [Google Scholar]
  165. Tedersoo L, Bahram M, Polme S, Koljalg U, Yorou NS, Wijesundera R, Villarreal Ruiz L, Vasco-Palacios AM, Thu PQ, Suija A, Smith ME, Sharp C, Saluveer E, Saitta A, Rosas M, Riit T, Ratkowsky D, Pritsch K, Poldmaa K, Piepenbring M, Phosri C, Peterson M, Parts K, Partel K, Otsing E, Nouhra E, Njouonkou AL, Nilsson RH, Morgado LN, Mayor J, May TW, Majuakim L, Lodge DJ, Lee SS, Larsson KH, Kohout P, Hosaka K, Hiiesalu I, Henkel TW, Harend H, Guo LD, Greslebin A, Grelet G, Geml J, Gates G, Dunstan W, Dunk C, Drenkhan R, Dearnaley J, De Kesel A, Dang T, Chen X, Buegger F, Brearley FQ, Bonito G, Anslan S, Abell S and Abarenkov K 2014. Global diversity and geography of soil fungi. Science 346:1256688. doi: 10.1126/science.1256688. [DOI] [PubMed] [Google Scholar]
  166. Thoms C, Ebel R and Proksch P 2006. Sequestration and possible role of dietary alkaloids in the sponge-feeding mollusk Tylodina perversa. Prog. Mol. Subcell. Biol 43:261–275. doi: 10.1007/978-3-540-30880-5_12. [DOI] [PubMed] [Google Scholar]
  167. Thornburg CC, Britt JR, Evans JR, Akee RK, Whitt JA, Trinh SK, Harris MJ, Thompson JR, Ewing TL, Shipley SM, Grothaus PG, Newman DJ, Schneider JP, Grkovic T and O’Keefe BR 2018. NCI Program for Natural Product Discovery: A publicly-accessible library of natural product fractions for high-throughput screening. ACS Chem. Biol 13:2484–2497. doi: 10.1021/acschembio.8b00389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Tran TD, Wilson BAP, Henrich CJ, Staudt LM, Krumpe LRH, Smith EA, King J, Wendt KL, Stchigel AM, Miller AN, Cichewicz RH, O’Keefe BR and Gustafson KR 2019. Secondary metabolites from the fungus Dictyosporium sp. and their MALT1 inhibitory activities. J. Nat. Prod doi: 10.1021/acs.jnatprod.8b00871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Tseng MH, Chou CH, Chen YM and Kuo YH 2001. Allelopathic prenylflavanones from the fallen leaves of Macaranga tanarius. J. Nat. Prod 64:827–828. doi: 10.1021/np0100338. [DOI] [PubMed] [Google Scholar]
  170. Uemura D, Takahashi K, Yamamoto T, Katayama C, Tanaka J, Okumura Y and Hirata Y 1985. Norhalichondrin A: An antitumor polyether macrolide from a marine sponge. J. Am. Chem. Soc 107:4796–4798. doi: 10.1021/ja00302a042. [DOI] [Google Scholar]
  171. Urban S, Brkljaca R, Hoshino M, Lee S and Fujita M 2016. Determination of the absolute configuration of the pseudo-symmetric natural product elatenyne by the crystalline sponge method. Angew. Chem. Int. Ed. Engl 55:2678–2682. doi: 10.1002/anie.201509761. [DOI] [PubMed] [Google Scholar]
  172. Wada N, Kersten RD, Iwai T, Lee S, Sakurai F, Kikuchi T, Fujita D, Fujita M and Weng JK 2018. Crystalline-sponge-based structural analysis of crude natural product extracts. Angew. Chem. Int. Ed. Engl 57:3671–3675. doi: 10.1002/anie.201713219. [DOI] [PubMed] [Google Scholar]
  173. Wagenaar MM 2008. Pre-fractionated microbial samples--the second generation natural products library at Wyeth. Molecules 13:1406–1426. doi: 10.3390/molecules13061406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Wall ME, Wani MC, Brown DM, Fullas F, Olwald JB, Josephson FF, Thornton NM, Pezzuto JM, Beecher CWW, Farnsworth NR, Cordell GA and Kinghorn AD 1996. Effect of tannins on screening of plant extracts for enzyme inhibitory activity and techniques for their removal. Phytomedicine 3:281–285. doi: 10.1016/S0944-7113(96)80067-5. [DOI] [PubMed] [Google Scholar]
  175. Walsh CT 2015. A chemocentric view of the natural product inventory. Nat. Chem. Biol 11:620–624. doi: 10.1038/nchembio.1894. [DOI] [PubMed] [Google Scholar]
  176. Wang M, Carver JJ, Phelan VV, Sanchez LM, Garg N, Peng Y, Nguyen DD, Watrous J, Kapono CA, Luzzatto-Knaan T, Porto C, Bouslimani A, Melnik AV, Meehan MJ, Liu WT, Crusemann M, Boudreau PD, Esquenazi E, Sandoval-Calderon M, Kersten RD, Pace LA, Quinn RA, Duncan KR, Hsu CC, Floros DJ, Gavilan RG, Kleigrewe K, Northen T, Dutton RJ, Parrot D, Carlson EE, Aigle B, Michelsen CF, Jelsbak L, Sohlenkamp C, Pevzner P, Edlund A, McLean J, Piel J, Murphy BT, Gerwick L, Liaw CC, Yang YL, Humpf HU, Maansson M, Keyzers RA, Sims AC, Johnson AR, Sidebottom AM, Sedio BE, Klitgaard A, Larson CB, Boya PC, Torres-Mendoza D, Gonzalez DJ, Silva DB, Marques LM, Demarque DP, Pociute E, O’Neill EC, Briand E, Helfrich EJ, Granatosky EA, Glukhov E, Ryffel F, Houson H, Mohimani H, Kharbush JJ, Zeng Y, Vorholt JA, Kurita KL, Charusanti P, McPhail KL, Nielsen KF, Vuong L, Elfeki M, Traxler MF, Engene N, Koyama N, Vining OB, Baric R, Silva RR, Mascuch SJ, Tomasi S, Jenkins S, Macherla V, Hoffman T, Agarwal V, Williams PG, Dai J, Neupane R, Gurr J, Rodriguez AM, Lamsa A, Zhang C, Dorrestein K, Duggan BM, Almaliti J, Allard PM, Phapale P, Nothias LF, Alexandrov T, Litaudon M, Wolfender JL, Kyle JE, Metz TO, Peryea T, Nguyen DT, VanLeer D, Shinn P, Jadhav A, Muller R, Waters KM, Shi W, Liu X, Zhang L, Knight R, Jensen PR, Palsson BO, Pogliano K, Linington RG, Gutierrez M, Lopes NP, Gerwick WH, Moore BS, Dorrestein PC and Bandeira N 2016. Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking. Nat. Biotechnol 34:828–837. doi: 10.1038/nbt.3597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Wang Y, Habgood GJ, Christ WJ, Kishi Y, Littlefield BA and Yu MJ 2000. Structure-activity relationships of halichondrin B analogues: Modifications at C.30-C.38. Bioorg. Med. Chem. Lett 10:1029–1032. doi: 10.1016/S0960-894X(00)00150-5. [DOI] [PubMed] [Google Scholar]
  178. Wani MC, Taylor HL, Wall ME, Coggon P and McPhail AT 1971. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J. Am. Chem. Soc 93:2325–2327. doi: 10.1021/ja00738a045. [DOI] [PubMed] [Google Scholar]
  179. Wani MC and Wall ME 1969. Plant antitumor agents. II. The structure of two new alkaloids from Camptotheca acuminata. J. Org. Chem 34:1364–1367. doi: 10.1021/jo01257a036. [DOI] [Google Scholar]
  180. Weinstein JN, Kohn KW, Grever MR, Viswanadhan VN, Rubinstein LV, Monks AP, Scudiero DA, Welch L, Koutsoukos AD, Chiausa AJ and Paull KD 1992. Neural computing in cancer drug development: Predicting mechanism of action. Science 258:447–451. doi: 10.1126/science.1411538. [DOI] [PubMed] [Google Scholar]
  181. Weinstein JN, Myers TG, O’Connor PM, Friend SH, Fornace AJ, Kohn KW, Fojo T, Bates SE, Rubinstein LV, Anderson NL, Buolamwini JK, van Osdol WW, Monks AP, Scudiero DA, Sausville EA, Zaharevitz DW, Bunow B, Viswanadhan VN, Johnson GS, Wittes RE and Paull KD 1997. An information-intensive approach to the molecular pharmacology of cancer. Science 275:343–349. doi: 10.1126/science.275.5298.343. [DOI] [PubMed] [Google Scholar]
  182. Wender PA, Hardman CT, Ho S, Jeffreys MS, Maclaren JK, Quiroz RV, Ryckbosch SM, Shimizu AJ, Sloane JL and Stevens MC 2017. Scalable synthesis of bryostatin 1 and analogs, adjuvant leads against latent HIV. Science 358:218–223. doi: 10.1126/science.aan7969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Williams JE 2001. Review of antiviral and immunomodulating properties of plants of the Peruvian rainforest with a particular emphasis on Una de Gato and Sangre de Grado. Altern. Med. Rev 6:567–579. doi: 0. [PubMed] [Google Scholar]
  184. Willot M, Radtke L, Könning D, Fröhlich R, Gessner VH, Strohmann C and Christmann M 2009. Total synthesis and absolute configuration of the guaiane sesquiterpene englerin A. Angew. Chem. Int. Ed. Engl 48:9105–9108. doi: 10.1002/anie.200905032. [DOI] [PubMed] [Google Scholar]
  185. Willstätter R and Müller W 1898. Ketones of the tropine group. XII. Constitution of ecgonine. Ber. Deutsch. Chem. Ges 31:2655–2669. doi: 0. [Google Scholar]
  186. Wosikowski K, Schuurhuis D, Johnson K, Paull KD, Myers TG, Weinstein JN and Bates SE 1997. Identification of epidermal growth factor receptor and c-erbB2 pathway inhibitors by correlation with gene expression patterns. J. Natl. Cancer Inst 89:1505–1515. doi: 10.1093/jnci/89.20.1505. [DOI] [PubMed] [Google Scholar]
  187. Wu Z, Zhao S, Fash DM, Li Z, Chain WJ and Beutler JA 2017. Englerins: A comprehensive review. J. Nat. Prod 80:771–781. doi: 10.1021/acs.jnatprod.6b01167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wynberg R 2004. Rhetoric, realism and benefit-sharing: Use of traditional knowledge of Hoodia species in the development of an appetite supressant. J. World Intellectual Property 7:851–876. doi: 10.1111/j.1747-1796.2004.tb00231.x. [DOI] [Google Scholar]
  189. Xu F, Nazari B, Moon K, Bushin LB and Seyedsayamdost MR 2017. Discovery of a cryptic antifungal compound from Streptomyces albus J1074 using high-throughput elicitor screens. J. Am. Chem. Soc 139:9203–9212. doi: 10.1021/jacs.7b02716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Xu Z, Metsä-Ketelä M and Hertweck C 2009. Ketosynthase III as a gateway to engineering the biosynthesis of antitumoral benastatin derivatives. J. Biotechnol 140:107–113. doi: 10.1016/j.jbiotec.2008.10.013. [DOI] [PubMed] [Google Scholar]
  191. Yin X, Zhou J, Jie C, Xing D and Zhang Y 2004. Anticancer activity and mechanism of Scutellaria barbata extract on human lung cancer cell line A549. Life Sci. 75:2233–2244. doi: 10.1016/j.lfs.2004.05.015. [DOI] [PubMed] [Google Scholar]
  192. Yoshioka S, Inokuma Y, Hoshino M, Sato T and Fujita M 2015. Absolute structure determination of compounds with axial and planar chirality using the crystalline sponge method. Chem. Sci 6:3765–3768. doi: 10.1039/c5sc01681a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Yu MJ, Zheng W, Seletsky BM, Littlefield BA and Kishi Y 2011. Case history: Discovery of eribulin (Halaven TM), a halichondrin B analogue that prolongs overall survival in patients with metastatic breast cancer Annual Reports in Medicinal Chemistry. (Macor JE, eds). pp. 227–241. Elsevier, Amsterdam. [Google Scholar]
  194. Zan J, Li Z, Tianero MD, Davis J, Hill RT and Donia MS 2019. A microbial factory for defensive kahalalides in a tripartite marine symbiosis. Science 364:1056. doi: 10.1126/science.aaw6732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Zani CL and Carroll AR 2017. Database for rapid dereplication of known natural products using data from MS and fast NMR experiments. J. Nat. Prod 80:1758–1766. doi: 10.1021/acs.jnatprod.6b01093. [DOI] [PubMed] [Google Scholar]
  196. Zhang C, Idelbayev Y, Roberts N, Tao Y, Nannapaneni Y, Duggan BM, Min J, Lin EC, Gerwick EC, Cottrell GW and Gerwick WH 2017. Small molecule accurate recognition technology (SMART) to enhance natural products research. Sci.Rep 7:14243. doi: 10.1038/s41598-017-13923-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Zhang H, Wang Y and Pfeifer BA 2008. Bacterial hosts for natural product production. Mol. Pharmacol 5:212–225. doi: 10.1021/mp7001329. [DOI] [PubMed] [Google Scholar]
  198. Zheng WJ, DeMattei JA, Wu JP, Duan JJW, Cook LR, Oinuma H and Kishi Y 1996. Complete relative stereochemistry of maitotoxin. J. Am. Chem. Soc 118:7946–7968. doi: 10.1021/ja961230+. [DOI] [Google Scholar]

RESOURCES