文档库 最新最全的文档下载
当前位置:文档库 › Natural product discovery past, present, and future

Natural product discovery past, present, and future

Natural product discovery past, present, and future
Natural product discovery past, present, and future

Natural product discovery: past, present, and future

Leonard Katz 1 · Richard H. Baltz 2

Received: 13 November 2015 / Accepted: 14 December 2015 / Published online: 6 January 2016 ? Society for Industrial Microbiology and Biotechnology 2016

precipitous drop in cost for genome sequencing, it is now feasible to sequence thousands of actinomycete genomes to identify the “biosynthetic dark matter” as sources for the dis-covery of new and novel secondary metabolites. Advances in bioinformatics, mass spectrometry, proteomics, transcrip-tomics, metabolomics and gene expression are driving the new field of microbial genome mining for applications in natural product discovery and development.

Keywords Actinomycete · Antibiotic · Discovery · Genome mining · Natural product · Fungi · Secondary metabolite · Streptomyces

Introduction

Natural products (NPs) represent a large family of diverse chemical entities with a wide variety of biological activi-ties that have found multiple uses, notably in human and veterinary medicine and in agriculture [26, 36, 47, 85, 86]. They originate from bacterial, fungal, plant, and marine ani-mal sources. The bacterial and fungal NPs described in this review are products of “secondary metabolism”: molecules that are not required for survival of the host under laboratory conditions, but which undoubtedly provide some advantage to the host in its native environment. Natural macromol-ecules (DNA, RNA, and protein), their building blocks and precursors, as well as intermediates of primary metabolism, are typically excluded from the working definition of NPs.NP discovery has been driven by the findings that they are important and valuable agents: pharmaceuticals, her-bicides, insecticides, etc. Since the discovery of penicillin more than 75 years ago, >23,000 NPs have been charac-terized, the large majority of which are produced by bacte-ria, mainly by the family Actinomycetaceae [25]. After the

Abstract Microorganisms have provided abundant sources of natural products which have been developed as com-mercial products for human medicine, animal health, and plant crop protection. In the early years of natural product discovery from microorganisms (The Golden Age), new antibiotics were found with relative ease from low-through-put fermentation and whole cell screening methods. Later, molecular genetic and medicinal chemistry approaches were applied to modify and improve the activities of important chemical scaffolds, and more sophisticated screening meth-ods were directed at target disease states. In the 1990s, the pharmaceutical industry moved to high-throughput screen-ing of synthetic chemical libraries against many potential therapeutic targets, including new targets identified from the human genome sequencing project, largely to the exclusion of natural products, and discovery rates dropped dramati-cally. Nonetheless, natural products continued to provide key scaffolds for drug development. In the current millennium, it was discovered from genome sequencing that microbes with large genomes have the capacity to produce about ten times as many secondary metabolites as was previously recog-nized. Indeed, the most gifted actinomycetes have the capac-ity to produce around 30–50 secondary metabolites. With the

Special Issue: Natural Product Discovery and Development in the Genomic Era. Dedicated to Professor Satoshi ōmura for his numerous contributions to the field of natural products. * Richard H. Baltz rbaltz923@https://www.wendangku.net/doc/417127319.html,

1

Synthetic Biology Engineering Research Center, University of California-Berkeley, 5885 Hollis St. 4th Floor, Emeryville, CA 94608, USA

2

CognoGen Biotechnology Consulting, 7636 Andora Drive, Sarasota, FL 34238, USA

discovery of streptomycin by Selman Waksman and associ-

ates at Rutgers University in the 1940s, large NP discovery

efforts began in pharmaceutical companies, mainly in the

United States, Europe and Japan, and modest efforts fol-

lowed in isolated academic laboratories worldwide. Nota-

ble exceptions include the academic laboratories of Satoshi ōmura at the Kitasato Institute (now Kitasato University) in Tokyo, the late Hamao Umezawa at the Institute of Anti-

biotics in Tokyo, the late Hans Z?hner at the University of

Tübingen, and Hans Reichenbach at the Helmholtz-Zen-

trum für Infektionsforschung in Braunschweig. Working

independently, and in collaboration with others, these labo-

ratories identified more than 1000 novel NPs.

What has fascinated NP scientists is the chemical diver-

sity and complexity of NPs, examples of which are dis-

played in the compounds shown in Figs. 1, 2, 3 and 4. Not

only is there a large range of molecular weights (coumerin,

146 Da to daptomycin, 1620 Da), but many of the com-

pounds contain a significant number of stereo-specific car-

bon centers. The stereo-complexity of these molecules has

attracted synthetic chemists interested in developing routes

and reactions for total chemical synthesis, and biochem-

ists and molecular biologists interested in understanding

the enzymology, genetics, and regulation of biosyntheses.

Microbiologists interested in the discovery of microor-

ganisms that produce novel molecules and novel activities

were needed to isolate and identify the producing micro-

organisms and to test and quantify the antibacterial or anti-

fungal activities. Cell biologists tested for anti-cancer and

other pharmacological activities. The commercial require-

ment for high volume production attracted chemical engi-

neers, fermentation microbiologists and geneticists, and

isolation and analytical chemists. Organic chemists were

attracted to pharmaceutical and biotechnology companies

to develop semi-synthetic derivatives. This family of aca-

demic and industrial scientists created the field of natural

product research.

This review covers the strategies and methodologies

employed from the 1940s through the present time to dis-

cover new NPs. We describe the screening for activity

approaches employed for the first 30 years of NP discovery,

highlighting some of the important molecules discovered

during “The Golden Age”. We then discuss how the under-

standing of NP biosynthesis changed the approaches for NP

discovery. For example, sequencing efforts demonstrated

that entire NP biosynthetic gene sets were clustered, and

allowed the use of genetic probes for the search for new

NPs, enabled the mixing, matching, and substitution of NP

biosynthesis genes to generate novel “hybrid” NPs, as well

as novel derivatives that could not be produced by synthetic

chemistry. We end with a discussion of the impact of the

development of inexpensive, high-throughput Next Gen-

eration Sequencing and advanced bioinformatics tools on the genomic approaches being taken for present and future NP discovery, focusing on genome mining and silent gene activation. The history of NP discovery, as we describe it, is segmented into three overlapping periods, 1940s–1970s, 1970s–2000s, and 2000 and beyond. These periods were chosen not because there were discrete breaks in the dis-covery process but because significant advances during these eras dramatically impacted the strategies employed to discover new NPs.

This review is not intended to be comprehensive, but to provide a basic understanding of the history of NP discov-ery without a particular emphasis on molecule type or med-ical field. Because of their overall value as drugs, however, which heavily skewed NP discovery efforts in pharma-ceutical companies towards specific medical applications during the first 30 years of effort, we present a brief sum-mary of the medical uses of NPs. Finally, because we both participated in one or more aspects of NP discovery over time, in addition to citing published material, we also use unpublished, personal accounts of our experiences in this perspective. For comprehensive reviews of the impact of NPs on medicine, readers are guided to other reviews [36, 85, 86]. For insider accounts on NP discovery and develop-ment programs in the Pharmaceutical Industry, readers are referred to Feature Articles in SIM News [8, 112, 127]. Natural products as drugs

As of 2013, 1453 new chemical entities (NCEs) have been approved by the US Food and Drug Administration, of which ca. 40 % are NPs or NP-inspired (semi-synthetic NP derivatives, synthetic compounds based on NP pharmaco-phores, or NP mimics) [36, 68]. In the past 30 years, the percentage of NP or NP-inspired NCEs has risen to approx-imately 50 % of the total, and to ca. 74 % in the anti-tumor arena [26, 47]. During the period 2008–2013, 25 NP or NP-derived drugs were approved for use. Here we present a brief summary of the many pharmaceutical applications of NPs or NP-inspired compounds and a short overview of their mechanisms of action or molecular targets. The struc-tures of the NPs highlighted in bold are shown in Figs. 1, 2, 3 and 4.

The most prominent medicinal use of NPs has been in the area of anti-infectives, notably anti-bacterial therapy (Fig. 1). Dozens of NPs, and their second- and third-gener-ation semisynthetic derivatives have been, and continue to be used to treat Gram-positive or Gram-negative bacterial infections in humans and animals. Briefly, azithromycin or clarithromycin, both semi-synthetic derivatives of the macrolide erythromycin, doxycycline, a second-genera-tion tetracycline, amoxicillin, a semi-synthetic penicillin derivative, and cephalexin, a derivative of cephalosporin

C, are all commonly used as outpatient treatments of bac-terial skin or respiratory infections. The macrolide tylosin and its semi-synthetic derivative tilmicosin are used to treat bacterial infections in animals. Macrolide antibacterial agents related to erythromycin or tylosin act as translation inhibitors by binding to the 50S ribosomal subunit, block-ing translation of the nascent polyptide chain. They are generally bacteriostatic for susceptible pathogens, but are bactericidal for Streptococcus pneumonia.Tetracycline

also blocks translation by binding to the 50S ribosomal Fig. 1 Structures of antibacterial natural products

subunit at a site distinct from the macrolides. Chloram?phenicol, a third ribosome binding agent is very active against Gram-negative bacteria, but is no longer used clini-cally, due to rare side effects. It binds to the A site of the 50S ribosome preventing the binding of aminoacyl-tRNA. The bactericidal penicillins and cephalosporins covalently bind to penicillin binding proteins in the bacterial cell wall and block cell wall synthesis, leading to exposure of the cell membrane to its osmotically unfavorable environment, resulting in cell lysis. Clavulanic acid, an inhibitor of sev-eral β-lactamases (which inactivate penicillins) is combined with the semi-synthetic penicillin analog amoxicillin in the compound Augmentin for use against penicillin-resist-ant pathogens. Glycopeptides, such as vancomycin and teicoplanin or their recently approved second-generation structural relatives (e.g., telavancin, dalbavancin, and orita-vancin) are used for Gram-positive infections that are not readily treated by macrolides and tetracyclines. The mem-brane active cyclic peptide daptomycin is used for hospi-tal-based difficult to treat Gram-positive infections, includ-ing methicillin-resistant Staphylococcus aureus (MRSA), and the penems (e.g., meripenem, derived from thienamy?cin) are used to treat infections caused by Gram-negative microorganisms. Natural penems are cephalosporin ana-logs. Aminoglycosides, such as tobramycin, are used in hospitalized patients carrying Gram-negative infections. They interact with mRNA in the 30S–50S ribosome junc-tion and cause mistranslation. Lipiarmycin (independently identified from three different actinomycetes, and also named clostomicin, tiacumicin and fidaxomycin), recently approved by the FDA, is effective against Clostridium difficile, the agent associated with severe colitis and diar-rhea. This macrolide does not bind ribosomes, but inhibits the β-subunit of RNA polymerase blocking transcription. Rifampicin, a semi-synthetic analog of rifamycin, also inhibits RNA polymerase and is used in combination with other agents, including streptomycin, for the treatment of tuberculosis.

Two different families of NPs are used to treat fungal infections in humans. Polyene macrolides such as ampho?tericin B (Fig. 2) and nystatin bind to ergosterol creat-ing holes in the fungal membrane and the echinocandins, such as caspofungin, inhibit the fungal cell wall form-ing enzyme glucan synthase. A large family of polyethers including monensin, narasin, salinomycin, and lasalocid are used in cattle feed as growth promoting agents or in chicken feed to prevent coccidiosis. The antimicrobial and antiparasidic effects of the polyethers are through their activities as ionophores, chelating mono- or divalent cati-ons. Ivermectin, a semi-synthetic derivative of avermectin, is active against parasitic worms in humans and animals. It is used to treat onchocerciasis (river blindness) and lym-phatic filariasis (elephantiais) in humans. Artemisinin is used to treat the malaria causing parasite Plasmodium fal-ciparum. Artemisinin is derived from a plant, but is now produced by the yeast Saccharomyces cerevisiae, based on a successful synthetic biology approach to heterologously

express the NP pathway. Spinosad, an antiparasitic and Fig. 2 Structures of antifungal and antiparasitic natural products

Fig. 3 Structures of anti-tumor natural products

insecticidal agent comprised of a mixture of the polyke-tides spinosyn A and spinosyn D, is approved for the topi-cal treatment of head lice in humans, and as an important insecticide active against a wide range of lepidopterous and dipterous insect pests in agriculture.

A large number of NPs or NP-inspired agents have been found to exhibit anti-tumor activity [26, 47]. Approved drugs from bacterial sources include the anthracycline mitomycin C (Fig. 3) and the mixed polyketide-nonribo-somal peptide (PK-NRP) bleomycin, both of which act by causing breaks in DNA, though via different chemical mechanisms; the di-cyclic peptide actinomycin D, a tran-scription inhibitor; doxorubicin, a semi-synthetic derivi-ative of the anthracycline polyketide daunorubicin, that acts by intercalating into DNA and blocking the enzyme topisomerase II; and romidepsin, a histone deacetylase inhibitor. Anti-tumor terpenes from plant sources include vincristine, vinblastine and paclitaxel. These agents block mitosis by binding to microtubules. Other anti-tumor agents from plants are the topoisomerase II inhibitor etopo-side, a semi-synthetic agent related to podophyllotoxin; the topoisomerase I inhibitor camptothecin, a quinoline alkyloid, as well as its water-soluble semi-synthetic deriva-tive topotecan; and the recently approved translation inhibi-tor omacetaxine mepesuccinate, a natural ester of the alka-loid cephalotaxine. Other recently approved anti-cancer drugs include the tetrapeptide epoxyketone carfilzomib, a semi-synthetic derivative of epoxomicin, and eribulin, a macrocylic ketone analog of the NP halichondrin B. Salinosporamide A is a proteosome inhibitor not yet approved as an anti-cancer agent. Many NPs that exhibit anti-tumor activities are too cytotoxic to be administered as drugs by themselves. A number has been conjugated to monoclonal antibodies for the purpose of target-specific delivery. NPs present in approved antibody–drug-conju-gates (ADCs) include the ansamycin polyketide maytan?sine and the acylpeptide dolastatin. The formerly approved ADC containing calicheamicin, an enediyne polyketide was recently withdrawn due to severe toxic side effects. However, many more enediynes have been discovered by focused genome mining [101], thus providing a wide range of chemical warheads for potential drug development.

An important advance in the treatment of hypercholes-terolemia was the development of the class of NPs known as statins, e.g., compactin (Fig. 4), lovastatin, and semi-synthetic derivatives simvastatin, atorvastatin, pravasta-tin. These compounds inhibit the mammalian and fungal enzyme HMG-CoA reductase in the cholesterol biosynthe-sis pathway. They continue to be among the highest rev-enue generating pharmaceuticals worldwide. The bacterial polyketides rapamycin and FK506 (tacrolimus), and the fungal cyclic peptide cyclosporine A are immunosupres-sants that are widely used after organ transplantation to sup-press rejection. Fingolimod, a drug used to treat multiple sclerosis, was developed from the fungal compound myri?ocin

, an inhibitor of serine palmitoyltransferase, by using Fig. 4 Structures of natural products with various bioactivities

structure–activity studies to determine the pharmacophore. Canagliflozin, a semi-synthetic derivative of the flavanoid phlorizin, found in many plants, is used to treat type 2 dia-betes, and capsaicin, derived from chili peppers, is used topically in over-the-counter creams to reduce minor aches and pains. In addition, many plant-derived NPs, e.g., cou?marin, are using in food as flavorings or as neutraceuticals.

Many of the examples described above represent FDA approved NPs or NP-inspired compounds. More than 100 other NPs or NP-inspired synthetic or semi-synthetic deriva-tives originally obtained from bacterial, fungal, plant, and marine sources are currently in clinical trials as anti-infective, anti-cancer, or other pharmaceutical agents. For a comprehen-sive list of NP-based compounds currently in clinical trials, the reader is directed to the excellent review by Butler et al. [30].

Each of the NPs described exhibits a distinct mechanism of action on their respective targets, although compounds within a given structural class with the same pharmacological activ-ity share binding sites in a common target. Erythromycin and tylosin both bind to the 23S rRNA in domain V of the bacterial ribosome, but the third sugar on tylosin also binds to domain II of 23S rRNA. The study of drug mechanism of action has revealed that similar pharmacological activities can be obtained from compounds of dissimilar structure. For example, rapa-mycin and cyclosporine A are both immunosuppressants, but have different targets and mechanisms of action. Other com-pounds, such as tetracycline and daunorubicin, have high struc-tural similarity but exhibit different pharmacological effects: antibacterial via binding to ribosome and anti-tumor via DNA intercalation, respectively. Finally some compounds have more than one pharmacological effect. For example, in addition to its antibacterial activity, erythromycin exhibits binding to motilin receptor in the human gut and is used clinically for treatment of diabetic gastroparesis, as well as to stimulate gastrointesti-nal motor activity when needed in patients following surgery. Because erythromycin is a large compound, the pharmacoph-ores responsible for the different properties are composed of different sub-sets of atoms of the molecule. These examples illustrate the variety of binding targets and pharmacological effects NPs have and underscore not only the values they have brought as medicines, but also as agents that have been used to deepen our understanding of fundamental biology.

Table 1 presents a list of 100 important NPs from diverse microbial sources and with diverse biosynthetic mecha-nisms. Most have use in human or animal medicine, or in agriculture. Several are important research tools, which will be further discussed below.

Strategies and methodologies for NP discovery Over the past seven decades, natural product discovery has been in a process of evolution; strategies were relatively simple during the first 30 years, then grew more diverse and complex, accelerated by advancements in science and technology in the next two decades. Overall efforts in NP discovery have decelerated with the waning interest from the pharmaceutical industry over the past twenty-years. In more recent times, inexpensive microbial genome sequenc-ing has opened totally new strategies for secondary metab-olite drug discovery. We recount some of the strategies and approaches in the following sections.

The first 30 years (1940s–1970s): phenotypic screening

Although the development of penicillin for wide use dur-ing WWII demonstrated success at bringing a NP to mar-ket, it was the systematic screening of actinomycetes obtained from soil by the Waksman group at Rutgers in the 1940s leading to the discoveries of actinomycin, strep-tothricin, and, most notably, streptomycin, that prompted pharmaceutical companies to begin screening extracts of actinomycetes and fungi for activities primarily against pathogenic bacteria. The process required acquisition of microorganisms, fermentation, product isolation, and test-ing of fermentation broths and purified compounds against test organisms. For testing, companies used bioassays for phenotypic screening. In general, a phenotypic screen employs a cell line (bacteria, yeast, eukaryotic cells or tis-sues in culture, etc.) and a method to determine a response to an applied test compound, without prior knowledge of its mechanism of action. The easiest read-out is cell inhibition or death, and, in the first 30 years this was used to iden-tify more than 1000 NPs that had antibacterial or antifungal activity, including dozens that were ultimately approved as drugs. Early cancer drugs were also detected in a pheno-typic screen using stable cancer cell lines cultivated first in experimental animal models and later, in vitro. Acquisition of microorganisms

Acquisition of actinomycetes and fungi was facilitated by the abundant numbers of these microbes in soil. At Eli Lilly and Company, Abbott Laboratories, and other phar-maceutical companies, employees were encouraged to col-lect soil samples when they traveled around the world on business or vacation. At Abbott, a large map of the world was kept on the wall of the soil screening laboratory with more than 2000 pins stuck at the locations from which soils were collected. Every continent, and almost every country had at least one pin. In many cases, new microbes produc-ing important compounds could be tracked back to the originating soils. The most famous example involved Pro-fessor Satoshi ōmura of the Kitasato Institute collecting a

Table 1 Diverse natural products produced as secondary metabolites by bacteria or fungi

Secondary metabolite Biosynthetic origin Major use Producing organism Type of organism Pubmed citations a

Acarbose Glycoside Antidiabetic (HM)Actinoplanes sp.Actinomycete1878 Actinomycin NRPS Antitumor (HM)Streptomyces anulatus Actinomycete24,705 Actinorhodin PKS II Antibacterial (RT)Streptomyces coelicolor Actinomycete448 Adriamycin PKS II Antitumor (HM)Streptomyces peucetius Actinomycete59,850 Albomycin Peptidyl-nucleoside Antibacterial Streptomyces sp. ATCC

700974

Actinomycete130 Amphomycin NRPS Antibacterial Streptomyces canus Actinomycete97 Amphotericin B PKS I Antifungal (HM)Streptomyces nodosus Actinomycete20,386 Antimycin NRPS-PKS I Research tool; Piscicide Streptomyces (multiple

sp.)

Actinomycete4558

Apramycin Aminoglycoside Antibacterial (AH, RT)Streptoalloteicus hindu-

stanus

Actinomycete303

Ascomycin NRPS-PKS I Immunomodulator

(HM)Streptomyces hygro-

scopicus

Actinomycete255

Avermectin (Ivermectin)b PKS I Anthelmintic (HM, AH)Streptomyces avermitilis Actinomycete741 (6208)b Bialphos NRPS Herbicide (CP)Streptomyces viridochro-

mogenes

Actinomycete182

Bleomycin NRPS-PKS I Antitumor (HM)Streptomyces verticillus Actinomycete17,239 Calcimycin (A23187)PKS I Calcium ionophore (RT)Streptomyces chartru-

ensis

Actinomycete11,796 (16,702)

Candicidin PKS I Antifungal (HM)Streptomyces (multiple

sp.)

Actinomycete291 Capreomycin NRPS Antitubercular (HM)Saccharothrix mutabilis Actinomycete545 Carbomycin PKS I Antibacterial (HM)Streptomyces halstedii Actinomycete166 Cephalosporin NRPS Antibacterial (HM)Cephalosporium acre-

monium

Fungus45,337

Cephamycin NRPS Antibacterial (S)Streptomyces cla-

vuligerus

Actinomycete3462 Chloramphenicol Shikimate modification Antibacterial (HM)Streptomyces venezuelae Actinomycete38,609 Chlorobiocin Aminocoumarine Antibacterial (S)Strteptomyces roseo-

chromogenes

Actinomycete96

Chloroeremomycin

(oritavancin)

NRPS Antibacterial (HM)Amycolatopsis orientalis Actinomycete39 (247)

Clavulanic acid (aug-mentin)Other Antibacterial (HM)Streptomyces cla-

vuligerus

Actinomycete6348 (12,346)

Compactin PKS Cardiovascular (HM)Penicillium brevicom-

pactum

Fungus662 Coumermycin Aminocoumarin Antibacterial (RT)Streptomyces rishiriensis Actinomycete353 Cyclosporin A NRPS Immunomodulator

(HM)

Tolypocladium inflatum Fungus50,544

d-Cycloserine Other Antitubercular (HM)Streptomyces lavendulae Actinomycete3045 Daptomycin NRPS Antibacterial (HM)Streptomyces roseospo-

rus

Actinomycete2183 Dalbavancin NRPS Antibacterial (HM)Nonomuraea sp.Actinomycete235 Daunorubicin PKS II Antitumor (HM)Streptomyces peucetius Actinomycete53,337 Echinocandin NRPS Antifungal (HM)Aspergillus nidulans Fungus3079 Epothilone NRPS-PKS I Anti-tumor (HM)Sorangium cellulosum Myxobacteria971 Ergometrine Ergolin alkaloid Cardiovascular (HM)Claviceps purpurea Fungus2180 Erythromycin PKS I Antibacterial (HM)Saccharopolyspora

erythraea

Actinomycete31,179 Filipin PKS I Antifungal (RT)Streptomyces filipinensis Actinomycete1303 Fusidic acid Terpine Antibacterial (HM)Fusidium coccineus Fungus2167

Geldanamycin PKS I Antitumor (S)Streptomyces hygro-

scopicus

Actinomycete1627

Gentamicin Aminoglycoside Antibacterial (HM)Micromonospora pur-

purea

Actinomycete27,233 Gramicidin S NRPS Antibacterial (RT)Bacillus subtilis Bacillales3841 Hygromycin Aminoglycoside Antibacterial (AH)Streptomyces hygro-

scopicus

Actinomycete2092

Josamycin PKS I Antibacterial (HM)Streptomyces narbon-

ensis

Actinomycete676

Kanamycin (amikacin)Aminoglycoside Antibacterial (HM)Streptomyces kanamy-

ceticus

Actinomycete18,934 (8055) Kirromycin NRPS-PKS I Antibacterial (RT)Streptomyces collinus Actinomycete219 Lasalocid PKS I Coccidiostat (AH)Streptomyces lasaliensis Actinomycete875

Lincomycin (clindamy-cin)Other Antibacterial (HM)Streptomyces lincoln-

ensis

Actinomycete7702 (9958)

Lipiaramycin (fidax-omicin)PKS I Antibacterial (HM)Dactosporangium

aurantiacum

Actinomycete155 (264)

Lipstatin (Xenical)Fatty acyl-lactone Antiobesity (HM)Streptomyces toxitricini Actinomycete25 (1559) Lovastatin PKS Cardiovascular (HM)Asperigillus terreus Fungus10,266 Milbemycin PKS I Anti-parasidic (AH)Streptomyces hygro-

scopicus

Actinomycete736 Mithramycin PKS II Antitumor (HM)Streptomyces plicatus Actinomycete1342 Mitomycin C Quinone Antitumor (HM)Streptomyces lavendulae Actinomycete18,112 Moenomycin Phosphoglycolipid Antibacterial (AH)Streptomyces ghanaensis Actinomycete228 Monensin PKS I Coccidiostat (AH)Streptomyces cinnamon-

ensis

Actinomycete4675

Mycophenolic acid PKS Immunomodulator

(HM)Penicillium brevicom-

pactum

Fungus7109

Narasin PKS I Coccidiostat (AH)Streptomyces aureofa-

ciens

Actinomycete221

Natamycin (pimaracin)PKS I Antifungal (HM)Streptomyces (multiple

sp.)

Actinomycete817 (894) Neomycin Aminoglycoside Antibacterial (HM)Streptomyces fradiae Actinomycete13,737 Netropsin NRPS Antitumor (RT)Streptomyces ambofa-

ciens

Actinomycete719 Nikkomycin Peptidyl-nucleoside Antifungal Streptomyces tendae Actinomycete246

Nisin RiPP Food preservative Lactococcus lactis Lactobacillales1720 Nystatin PKS I Antifungal (HM)Streptomyces noursei Actinomycete4686 Oligomycin PKS I Toxin (RT)Streptomyces avermitilis Actinomycete4784 Oxytetracycline PKS II Antibacterial (HM)Streptomyces rimosus Actinomycete7379 Paclitaxel Isoprenoid Antitumor (HM)Several endophytic fungi Fungus27,268 Penicillin NRPS Antibacterial (HM)Penicillium crysogenum Fungus96,006 Phosphomycin Phosphone Antibacterial (HM)Streptomyces wedmo-

rensis

Actinomycete2410 Pleuromutalin Diterpine Antibacterial (HM)Clitopilus scyphoides Fungus161 Pneumocandin NRPS Antifungal (HM)Glarea lozoyensis Fungus77 Polymyxin (colistin)NRPS Antibacterial (HM)Paenibacillus polymyxa Bacillales9611 (4084) Polyoxin (D)Nucleoside Antifungal (CP)Streptomyces cacaoi Actinomycete120 Pristinamycin IA NRPS Antibacterial (HM)Streptomyces pristi-

naespiralis

Actinomycete521

Pristinamycin IIA NRPS-PKS I Antibacterial (HM)Streptomyces pristi-

naespiralis

Actinomycete131

soil sample from a golf course in Tokyo in the early 1970s from which his research group discovered avermectin from a new species of Streptomyces that he named S. avermiti-lis. Other examples include the erythromycin producer Saccharopolyspora erythraea, discovered in 1949 in the garden of a local physician in the Phillipines employed by Eli Lilly and Company [https://www.wendangku.net/doc/417127319.html,/1994/11/ medicine-philippines-who-really-discovered-erythromycin-1-an-inter-press-service-feature], the daptomycin producer Streptomyces roseosporus, tracked to a soil sample from

Ramoplanin NRPS Antibacterial (HM)Actinoplanes sp. ATCC

33076

Actinomycete155

Rapamycin NRPS-PKS I Immunomodulator

(HM)Streptomyces hygro-

scopicus

Actinomycete26,695

Rebeccamycin Alkaloid Antitumor Lechevalieria aerocolo-

negenes

Actinomycete128

Rifamycin (rifampicin)PKS I Antibacterial (HM)Amycolatopsis mediter-

ranei

Actinomycete19,004 (24,279) Ristocetin NRPS Antibacterial (RT)Amycolatopsis lurida Actinomycete2953 Salinomycin PKS I Coccidiostat (AH)Streptomyces albus Actinomycete624 Sinefungin Nucleoside Antifungal (RT)Streptomyces griseolus Actinomycete255 Spectinomycin Aminoglycoside Antibacterial (HM)Streptomyces spectabilis Actinomycete2408 Spinosyn (spinosad)PKS I Insecticidal (CP)Saccharopolyspora

spinosa

Actinomycete92 (548)

Spiramycin PKS I Antibacterial (HM)Streptomyces ambofa-

ciens

Actinomycete1367

Staurosporin (aglycone)Alkaloid Antitumor (S)Streptomyces stauro-

sporeus

Actinomycete10,116 Streptomycin Aminoglycoside Anti-tubercular (HM)Streptomyces griseus Actinomycete29,437 Streptothricin Aminoglycoside Antibacterial Streptomyces (multiple

species)

Actinomycete337

Streptozotocin Glucosamine-nitros-

ourea Antitumor (HM)Streptomyces acromo-

genes

Actinomycete24,436

Surfactin NRPS Surfactant Bacillus subtilis Bacillales595

Tacrolimus (FK-506)NRPS-PKS I Immunomodulator

(HM)Streptomyces tsukubae-

nsis

Actinomycete19,514 (19,841)

Teicoplanin NRPS Antibacterial (HM)Actinoplanes teichomy-

ceticus

Actinomycete3358

Tetracenomycin PKS II Antitumor Streptomyces glauce-

scens

Actinomycete101 Tetracycline PKS II Antibacterial (HM)Streptomyces rimosus Actinomycete35,863 Thienamycin Other Antibacterial (HM)Streptomyces cattleya Actinomycete324 Thiostrepton RiPP Antibacterial (AH, RT)Streptomyces azureus Actinomycete486 Tobramycin Aminoglycoside Antibacterial (HM)Streptoalloteicus hindu-

stanus

Actinomycete6636 Tunicamycin Nucleoside Antibacterial (RT)Streptomyces chartreusis Actinomycete4971

Tylosin PKS I Antibacterial (AH)Streptomyces fradiae Actinomycete1557 Undecylprodigiosin Other Antibacterial (RT)Streptomyces coelicolor Actinomycete161 Validamycin Glycoside Antifungal (CP)Streptomyces hygro-

scopicus

Actinomycete99

Viomycin NRPS Antibacterial (HM)Streptomyces sp. 11861Actinomycete831 Virginiamycin NRPS, NRPS-PKS I Antibacterial (AH)Streptomyces virginiae Actinomycete1372

AH animal health, CP crop protection, HM human medicine, PKS polyketide synthase, NRPS nonribosomal peptide synthetase, S scaffold for chemical semi-synthesis, RiPP ribosomally synthesized and post-translationally modified peptide, RT research tool

a PubMed citations on 8/31/2015

b Compounds of commercial products in parentheses correspond to citations in parentheses

Mount Ararat in Turkey [41], and the spinosad producer Saccharopolyspora spinosa, tracked to a defunct sugar mill rum still in the Virgin Islands [125]. The practice of collect-ing soil from foreign sources and bringing samples back to the home country went on unabated until countries signed the treaty known as the Convention on Biological Diversity (CBD) in the early 1990s. Until then, the countries from which the soil samples originated did not share in the reve-nues brought by the subsequent drugs that were developed. CBD ensured that revenues would return to these countries.

During the first three decades of antibiotic discovery in the pharmaceutical industry, many important NPs were dis-covered from microbial sources, many of which, or their second, third, or fourth generation semi-synthetic deriva-tives, are still in use today. Many of the important NPs listed in Table 1 were discovered in this timeframe. Thou-sands of new chemical entities (NCEs) were discovered from soil screening, but only a small number were devel-oped as drugs. Nevertheless, the success achieved from the commercial launch of a single compound was sufficient to keep the process going unchanged for more than 30 years, at least in the companies with which we are familiar. In general, soil screening was a low-throughput process which, as described below, was limited by the slow growth of the actinomycetes and fungi examined, and importantly by the amount of shaker space available to companies.

In the early years of screening, most companies focused on isolating and identifying Streptomyces as the major bac-terial genus to search for NCEs. As can be seen in Table 1, Streptomyces species yielded many important compounds in several antibiotic classes: macrolides (tylosin, spiramy-cin); aminoglycosides (neomycin, kanamycin,); β-lactams (cephamycin, carbapenems); tetracyclines (tetracycline, chlortetracycline, oxytetracycline); polyenes (candicidin, amphotericin B, nystatin); peptides (actinomycin); and chloramphenicol. However, three important antibiotics discovered by Eli Lilly and Company, erythromycin, van-comycin and tobramycin [8], were originally thought to be produced by Streptomyces species, but the producing actinomycetes were later reclassified as Saccharopolyspora erythraea, Amycolatopsis orientalis, and Streptoalloteicus hindustanus, respectively (Table 1). In the late 1950s, the US-based Schering Corp. collected a large group of soil actinomycetes classified as Micromonospora and began screening them for antibacterial activity [124, 127]. This genus yielded the aminoglycosides gentamicin (a fam-ily consisting of more than 5 congeners), sisomicin, the fortimicins, and sagamicin, the macrolides rosaramicin, juvenimicin, and the megalomicins, as well as the oligo-saccharide everninomicin. Only gentamicin was developed as an antibacterial drug. Lepetit (later Dow Lepetit then Biosearch Italia) began to screen rare actinomycetes in the 1960s. The company discovered the macrolide lipiarmycin from Dactosporangium (later developed as fidoxamicin by Optimer and Cubist), and the glycopeptides teichopla-nin and ramoplanin from different Actinoplanes species. It is now apparent from the early discoveries in the pharma-ceutical industry that in addition to the highly productive Streptomyces, other less abundant actinomycetes have con-tributed substantially to the wealth of important NPs dis-covered in the first 30 years.

Detection, fermentation and isolation of secondary metabolites

To determine if they produced active compounds, indi-vidual colonies isolated from agar plates were grown in small shake flask fermentations employing several media compositions, and in some cases under a range of tempera-tures and agitation speeds. Initial detection for antibacterial or antifungal activity involved assay of solvent extracted culture broths. Small amounts of extracted material were deposited on filter paper discs which were then placed on agar plates seeded with a susceptible test organism. Detec-tion was as simple as visualization of growth inhibition of test organisms after overnight incubation. Activities were quantified by standard MIC analyses employing panels of various pathogens. For more polar compounds that were not solvent extractable (e.g., peptidic or glycosidic agents), Schering used an agar plug assay. Small (2–3 cm diam-eter) plugs were removed from agar plates seeded with a test organism, allowing the administration of up to 5 mL of whole culture broths of soil isolates (Micromonospora strains). Detection of small zones of inhibition after over-night incubation was used to identify antibiotic producers. After the activities were identified, the active components were often separated by paper chromatography and assayed by bioautography: the chromatogram was placed on an agar plate seeded with a susceptible test strain; after overnight incubation, the plate was examined for zones of inhibition corresponding to the migration of the active ingredient. Bioautography was used to determine if there was more than a single active compound in the culture. After initial discovery, product titers were improved initially by fer-mentation optimization to produce enough material for iso-lation and evaluation. In some cases, the producing strain was subjected to mutagenesis and selection for product titer improvement at an early stage using chemical or physical mutagenic agents. Promising strains were then scaled in pilot plant fermenters to obtain greater quantities of mate-rial for isolation and structural characterization, and further activity evaluation. Because of the ease of collecting sam-ples and obtaining different colonies, each colony (strain) was typically given a single chance, as described above, to produce an active product, although many companies kept the isolated colonies as lyophilized spore suspensions in

their culture collections. Initial shake flask cultures were generally discarded. Because of the sheer volume of shake flasks and shaker capacity, companies could test only a limited number of different microbes per year. At Eli Lilly and Company, that number was about 35,000, translating to about 1,000,000 strains screened over three decades. [8, 11]. By the mid-1990s, when NP screening was halted, Abbott Laboratories had screened about 400,000 isolates (J. McAlpine, personal communication).

We estimate, for all companies, institutes, and research laboratories worldwide engaged in screening during the period from 1950 to 2000, that about 10–20 million isolates were screened. Undoubtedly, many of the more common actinomycete species were screened many times by many companies. From these, thousands of NCEs were identified and hundreds, either the NPs themselves or their deriva-tives, were developed as drugs. Although, as we describe briefly below, the methods to screen for pharmacological activities have changed dramatically over the last 50 years, the processes employed to isolate organisms from envi-ronmental samples, to grow them in liquid culture, and to extract and purify natural products have undergone only minor evolution. High-speed shakers capable of providing sufficient aeration to allow actinomycetes to grow in micro-titer plates [80] replaced the need for shake flask cultivation so that more isolates could be screened in less space, and major improvements in NMR [29] and MS [49] analyses have made structure determination of complex NPs easier and quicker. Large-scale fermentation has remained basi-cally unchanged.

The first 30 years of NP discovery followed the para-digm: (1) phenotypic screening, (2) compound isola-tion and structural characterization, (3) mode of action studies in some cases, (4) preclinical development, and if successful, (5) clinical development and commerciali-zation. Efforts were directed primarily to discover anti-bacterial and antifungal compounds. All of the NPs with antibacterial or antifungal activities discovered during this period, with a single exception, employed pheno-typic screens that used cell killing (or growth inhibition) as the read-out. Because Merck was interested in discov-ering agents specifically targeted to the biosynthesis of the cell, they developed a screen in which the read out was the conversion of bacteria to spheroplasts. This led to the discovery of fosfomycin [53, 110]. Moreover, with a single exception, all the antibacterials and antifun-gals discovered during this era that went on to be devel-oped as drugs were all NPs. Naladixic acid is based on the structure of the NP quinine, and was identified in a search for synthetic derivatives of anti-malarial drugs, but was found to have serendipitous antibacterial proper-ties. In the 1980s, a number of natural quinolones with antibacterial properties were discovered from the bacte-rium Pseudonocardia spp. CL3849, as well as various plant sources [50].

As early as the 1950s, companies became interested in expanding their screening efforts to find compounds for treatment of cancer. Anti-cancer agents were screened by observing the reduction of transplanted or experimentally induced tumors in rodents or by observing cytotoxic effects of tumor cell lines grown in culture. Whole animal tests were also used to distinguish antibacterial or antifungal activity from cytotoxic activity and were used to inform companies that antibacterial agents such as kirromycin or oligomycin, or the antifungal agent rapamycin, for exam-ple, were too toxic to be considered for drug development. On the other hand, the same assays were used to initially identify the anti-tumor activities of daunorubicin, mito-mycin C, bleomycin, and camptothecin (Fig. 1) [39, 46, 63, 122]. In the 1960s and 1970s, companies also started to become interested in finding NPs for treatment of heart disease, neurologic disorders, and metabolic diseases, but facile assays to screen for effective compounds in these therapeutic areas were not readily available. Sandoz (now Novartis) discovered cyclosporine A (Fig. 4) using an assay developed to test both immunosuppressive and anti-tumor activities in a single mouse. [55, 109]. Blood drawn from mice 7 days after they were immunized with sheep erythrocytes (antigen) agglutinated when the antigen was added to the test tube. To test for immunosuppressive activ-ity, the agent was given by intravenous administration to the mice at the time of inoculation with the antigen, and for a number of days thereafter. If the blood drawn at 7 days did not agglutinate in the presence of the antigen, the com-pound was said to have blocked the immune response–immunosuppressive. Because the same mice carried trans-planted tumors, antitumor activity could be measured by suppression of tumor growth after the test compound was administered. This expensive, low-throughput method was replaced by the mixed lymphocyte reaction (MLR) test in the 1980s, which is described below.

The second 30 years (1970s–2000s):

knowledge?based approaches

As mentioned above, other than some miniaturization of organism cultivation and improvements in NMR and MS analyses for structure determination, used extensively for dereplication purposes (to eliminate rediscovery of known compounds), the processes used to produce NPs for discovery during the second 30 years remained largely unchanged. New sources for NPs were sought during this period. The hallmark of the second 30 year period was the

great expansion in screening methodologies and strategies. Advancements in recombinant DNA and other technolo-gies enabled researchers to rapidly determine mechanism of action (MOA) of NPs, their semi-synthetic analogues, and synthetic compounds, and rapidly led to the develop-ment of biochemical or whole cell assays, collectively known as target-based screening. In addition, the growing understanding of cell cycle regulation, the role of recep-tors in biologic responses, the genetic bases of cancer, etc., along with improvements in cell culture methodology, led to the development of a large number of selective pheno-typic screens for agents active against cancers, various neu-rologic, metabolic, or cardiac disorders or abnormalities. Both target-based and phenotypic screening are too vast to be described comprehensively here, but are covered in a number of reviews [74, 81, 102, 118]. We present here a brief overview of some of the approaches taken, including a number of examples that led to successful development of NP-based drugs.

New sources of NPs

Phenotypic screening of soil samples for NPs continued in the 1970s and 1980s, but was expanded, as described below, beyond the search for antibacterial or antifungal drugs. In addition, new sources beyond soils were sought for NP discovery. This included marine environments, with a heavy focus on the discovery of novel anti-cancer agents. Samples collected by certified divers or sampling devices included bacteria, algae, and marine invertebrates (tuni-cates, corals, bryzoans, sea slugs, sponges, etc.), and the NPs extracted from the samples were tested in new anti-cancer screens described below.

Phenotypic screening

As described above, the early cancer drugs, mitomycin C, daunoribucin, and actinomycin (Fig. 3) were discovered using cancer cell lines transplanted into animals. Starting in the 1960s, companies and academic laboratories interested in anti-cancer agents began to produce stable cell lines that were differentiated for specific types of cancer (e.g., lung, breast, etc.) to screen fermentation broths, as well as their compound collections consisting of purified NPs, semi-synthetic and synthetic chemicals. In the 1980s, the US National Cancer Institute began to develop their own col-lection of stable, differentiated specific cancer cell lines, culminating in the collection known as NCI-60, a panel of 60 cell lines differentiated for breast, lung, colon, prostate, renal, ovarian, or CNS cancers, as well as those for leuke-mia, and melanoma, and set up a service to test investiga-tional compounds (including NPs) for efficacy (providing dose response curves) and specificity (IC50 determinations using the entire NCI-60 panel). For compounds discov-ered before the panel was in use, for example paclitaxel (Fig. 3), the results provided by NCI led to the develop-ment of the drug for use in breast, lung and ovarian cancer. The primary use of the NCI-60 panel today, however, is for screening (efficacy, specificity, and MOA) of new NPs, and other putative anti-cancer agents [104].

As of 2014, three compounds derived from NPs from marine invertebrates have been approved for treatment of various cancers. These include eribulin, a synthetic derivative of halichondrin B (Fig. 3), trabectidin, a semi-synthetic derivative of the NP safricin B, and PM-10450, a synthetic compound derived from jorumycin from a sea slug, and renieramycin J from a marine invertebrate. It is not yet known if these compounds are produced from the marine invertebrates themselves, of from yet to be identi-fied bacterial symbionts. A number of other semi-synthetic or synthetic derivatives, or antibody-conjugates of marine invertebrate NPs are in various stages of clinical trials. The compound salinosporamide A (Fig. 3), isolated from the marine bacterium Salinispora tropica is the single example to date of a potential anti-cancer NP from a marine bacte-rium that made it into clinical trials [30].

A second example of a phenotypic screen developed in the second period of NP discovery is the mixed lympho-cyte reaction (MLR), used to screen for immunosuppres-sive agents. In general when lymphocytes of two different allotypes are mixed, an immunologic response causes them to grow and divide. If one of the set of lymphocytes is uv-inactivated, it can serve as a stimulator for growth and divi-sion of the un-irradiated responder lymphocytes. Growth can be measured by following 3H-thymidine incorporation (into DNA) or by following OD570 in cells treated with the tetrazolium dye MTT (3-[4,5-dimethylthiozol-2-yl]-2,5-di-phenyltetrazoium bromide) [37, 82]. Immunosuppressive activity is detected by inhibition of growth of the responder cells. Separate cell-based assays are used to distinguish the immunosuppression from cytoxicity. Many versions of the MLR have been developed, from employing whole blood from different donors, to using splenocytes drawn from dif-ferent lines of mice [60]. Validation of the MLR employed the use of cyclosporin A (Fig. 4) to demonstrate immu-nosuppression. The MLR assay played a prominent role in the discovery of two new NPs currently approved as immunosuppressive agents, FK506 (tacrolimus) and rapa?mycin (Fig. 4). The immunosuppressive activity of FK506, produced by Streptomyces tsukubaensis, was detected at Fujisawa (now Astellas) in a primary screen using the MLR assay. Rapamycin, isolated from a strain of Streptomyces hygroscopicus collected on Easter Island, was discovered in the early 1970s by microbiologists at Ayerst Research Lab-oratories in an antifungal screen, but the compound proved to be too toxic to develop as an antifungal drug [123]. Two

years after the initial report of the antifungal activity, phar-macologists at Ayerst reported immunosuppressive activity demonstrating the complete prevention of experimentally induced immunogenic disorders in rats [75]. Work on the mode of action of rapamycin, including demonstration that at least one binding target of the drug differed from that of FK506 continued during the 1980s, but serious develop-ment of rapamycin as an immunosuppressive drug did not begin until the late 1990s, after direct comparisons of activ-ity between rapamycin and FK506 or cyclosporin A using the MLR assay were published [52].

Target?based approaches

In phenotype screens, the mode of action (i.e., molecular target) of the compound tested is not known or assumed. In contrast, target-based screening is based on the implica-tion or the validated understanding that interaction of tested compound with the designated target (usually an enzyme, enzyme complex, or a receptor) will result in the desired pharmacological effect. The screen is an assay that pro-vides a measurable read-out if the tested compound inter-acts specifically with the target. Target-based assays can be as simple as in vitro enzyme assays. Target-based assays can also employ genetically engineered yeast or bacteria in whole-cell survival read outs, similar to that used in pheno-typic screening.

Only a very small number of NPs were identified by using a specific enzyme assay and later developed as drugs. Two are described here: lovastatin and clavulanic acid. As early as the middle 1960s, the biochemical pathway of cho-lesterol biosynthesis was fairly well understood and the enzyme hydroxymethylglutaryl-CoA reductase (HMGR) was known to convert HMG-CoA to the intermediate mevalonate. Akira Endo at Sankyo developed a three step assay (two phenotypic screens and a target-based assay) using liver tissues extracted from rats to screen fermenta-tion broths to find NPs that specifically inhibited HMGR [43]. In the first screen, the broths were screened examin-ing the conversion of 14C-acetate to 14C-non-saponifiable lipids (e.g., cholesterol). Broths that exhibited inhibition were then screened in an assay that measured conversion of 3H-mevalonate to 3H-lipids. Broths that did not inhibit this conversion were then assayed for the conversion of 14C-HMG-CoA to 14C-mevalonate. Using this approach, Endo discovered the compound ML-236B, later named compactin (Fig. 4), from the broth of a strain of Penicil-lium citrinum that was isolated from a rice sample collected from a vendor in Kyoto. Compactin was not developed as a drug, however. Using screening strategies analogous to that used to discover compactin, Endo and a group at Merck independently discovered lovastatin, a methylated analog of compactin, from two different fungi, Monascus ruber and Aspergillus terreus, respectively [2, 42]. Merck developed lovastatin to become the first in class of the statins successfully used to treat hypercholesteremia. Sec-ond-generation synthetic derivatives of compactin and lov-astatin, known under the trade names Pravachol?, Zocor?, and Lipitor? are among the world’s largest selling drugs. In retrospect, the contributions that Endo brought to human health with the discovery of compactin may not have been adequately recognized.

Clavulanic acid was discovered by Beecham [98] in a culture broth of a cephamycin producing strain of Strep-tomyces clavuligerus first described by Lilly scientists in 1971 [8, 83]. Although clavulanic acid was not active enough to be considered for development as an antibacte-rial agent, it was later shown to be an effective competi-tive inhibitor of a number of β-lactamases which, by as early as the 1950s, were known to be mediators of clinical resistance of Gram positive and Gram negative pathogens to penicillin and other β-lactam antibiotics [95]. Clavu-lanic acid has been combined with amoxicillin, a semi-syn-thetic derivative of penicillin, in the drug Augmentin? to widen the spectrum of pathogens susceptible to antibiotic treatment.

During the second 30 year period, discovery of new NPs from screening continued. We cannot accurately state the number of NCEs discovered during this period, but the number of patents filed world-wide exceeded 1300 [70]. Yet less than two dozen novel NPs, their-semi-synthetic or completely synthetic derivatives that were discovered dur-ing this period were developed as drugs [114]. In contrast to the first 30 year period, which gave rise to the introduc-tion of dozens of antibacterials NPs into clinical practice, the only novel anti-bacterial NP discovered during the sec-ond 30 year period that went through clinical development was daptomycin; the echinocandins represent the only class of novel antifungal NPs discovered during this period that were later approved as drugs. On the other hand, numerous 2nd, 3rd, and in the case of the cephalosporins, 4th genera-tion semi- or completely synthetic derivatives of antibac-terial NPs, representing many chemical classes, were suc-cessfully developed during this period.

During the 1990s, combinatorial chemistry was devel-oped that could give rise to thousands of unique com-pounds from hundreds of chemical scaffolds. Most phar-maceutical companies either purchased or developed their own ‘combi-chem’ libraries that numbered in the 5 × 105 to 4 × 106 range. In addition, advances in robotics together with the development of receptor-binding or enzyme-based assays with facile read-outs enabled companies to run these libraries through multiple high-throughput screens (HTS) simultaneously. To handle the large numbers, mixtures were made containing known compounds and screened; mixtures yielding “hits” were deconvoluted in subsequent

screening rounds. In contrast, NP preparation of broths could not be scaled up practically and, hence, could not keep pace with the generation of known synthetic chemi-cals. The prospect of screening endless numbers of known compounds rapidly versus the low-throughput phenotypic screens of small numbers of broths that may or may not have contained NPs was too enticing for management in pharmaceutical companies to pass up. By the mid-1990s, most pharmaceutical companies in the US and Europe stopped screening natural products. The exception was Novartis, which continues NP screening [102].

During the period between the mid-1990s to the mid-2000s, millions of compounds were screened against hun-dreds of targets in areas relating to metabolic diseases, cancer, neurobiology, cardiovascular, immunology, and infectious disease. More compounds were screened in this period than in the preceding 60 years of drug discovery, yet this effort returned very few quality compounds, and to date has produced no approved drugs. GSK ran a library of upwards of 500,000 synthetic compounds through 70 antibacterial screens on targets from Streptococcus pneu-monia. All the targets were validated as essential proteins involved in macromolecular biosynthesis, including targets for which there were known NP antibiotics: MurA, fosfo-mycin; FabI/FabH, cerulenin, thiolactomycin; PBP2, peni-cillins, cepahlosporins. All of the screens either returned no hits, or hits that were not turned into leads. The same 500,000 compound library was also run through whole cell antibacterial screens using E. coli and Staphyloccocus aureus, again resulting in no useful leads [96]. Mutasynthesis and precursor?directed biosynthesis Mutational biosynthesis, commonly referred to as muta-synthesis, is the production of novel NPs through the incorporation of analogs of NP intermediates into biosyn-thetic pathways. This approach, coined by Kenneth Rine-hart, was started in the late 1960s in his laboratory, and used extensively throughout the 1970s and 1980s. Feed-ing streptamine or epistreptamine, analogs of the normal precursor 2-deoxystreptamine, to mutants of Strepomyces fradiae blocked in neomycin biosynthesis, caused produc-tion of neomycin analogs [103]. Similar mutasynthesis programs were used to produce analogs of novobiocin, the nucleoside antibiotic nikkomycin, as well as other NPs [129]. The early studies were undertaken before full knowledge of the biochemical pathways became available. Understanding the steps in secondary metabolite biosynthe-sis, coupled with mutagenesis to block specific steps has enabled a related approach termed product-directed bio-conversion [10]. An example is the approach was taken at Eli Lilly and Company in the 1980s to generate macrolide antibiotics related to tylosin. Macrocin, the immediate precursor to tylosin, was produced by a mutant blocked in terminal O-methylation generated by N-methyl-N’-nitro-N-nitrosoguinadine (MNNG) mutagenesis, affording a fer-mentation process to overproduce macrocin [22]. Purified macrocin was then fed to Streptomyces thermotolerans, a strain that expresses 4″-mycarosyl isovaleryl-CoA trans-ferase, leading to the biosynthesis of 4″-isovalerylmacro-sin, a highly active antibiotic that was evaluated as an ani-mal health product by Lilly Elanco [121]. Although Lilly Elanco chose not to develop this compound, other compa-nies currently manufacture a related compound, 3-acetyl-4″-isovaleryltylosin (AIV or Tylvalosin), for treatment of Mycoplasma gallisepticum air sac infections in chickens, by feeding tylosin to Streptomyces thermotolerans for bio-conversion [135].

Other mutants blocked in tylosin biosynthesis produced high levels of other precursors or shunt metabolites which served as starting materials for semi-synthesis. One mutant produced desmycosin, which lacked a single sugar resi-due. Desmycosin was used as a scaffold for semi-synthesis culminating in the discovery of tilmicosin [35], marketed by Lilly Elanco as Micotil?. Semi-synthesis has also been used to generate a second generation derivative of spino-sad (Fig. 2), a glycosylated type I polyketide insecticide marketed by Dow AgroSciences for plant crop protec-tion. MNNG-induced mutants of Saccharopolyspora spi-nosa blocked in spinosad biosynthesis were isolated and evaluated for insecticidal activities [69, 125]. One of the mutants was defective in the O-methylation of the 3′-OH of the ramnosyl moiety and accumulated high levels of fac-tors J and L, the starting materials for the semi-synthesis of spinetoram, which is marketed as Radiant? by Dow Agro-Sciences [45, 108].

Precursor-directed biosynthesis also employs the addi-tion of specific compounds to the growth medium that ulti-mately get incorporated into the pathway resulting in the biosynthesis of a novel NP. Unlike mutasynthesis, however, the compound supplied does not replace an intermediate in the pathway, but a precursor, hence mutants employed are not in the biochemical pathway per se. The best example of precursor-directed biosynthesis is the production of the anthelminthic compound doramectin. By the early 1980s, it was well understood that the avemectins employed the precursors isobutyryl-CoA (e.g., avemectin 1B; Fig. 2) or 2-methylbutyryl-CoA to initiate their biosyntheses in S. avermitilis. In the 1990s, the Pfizer group identified the set of bkd genes in S. avermitilis that were respon-sible for the breakdown of branched-chain amino acids valine, isoleucine, and leucine that resulted in the produc-tion of isobutyryl-CoA and 2-methylbutyryl-CoA, as well as isovaleryl-CoA. After disrupting this pathway, they found that numerous carboxylic acids could be introduced into the “starting” position of avermectin, giving rise to a

variety of novel analogs [38]. Among them was the com-pound named doramectin, which carries cyclohexane in place of the isobutyate or 2-methylbutyrate side chain. Ser-endipitously, the S. avermitilis avermectin producing strain contains one or more acyl-CoA synthetases that have broad substrate specificities which, upon direct feeding of free acids, enable them to generate a variety of acyl-CoAs that can initiate avermectin biosynthesis. A similar approach to changing the structure of the starter employed in rapamy-cin biosynthesis was attempted. The dihydroxycyclohexyl starter unit of rapamycin was replaced with a hydroxycy-clohexyl- or hydroxycyclohepatanyl-unit when the corre-sponding carboxylic acids were fed to a rapK mutant of the rapamycin producer, Streptomyces hygroscopicus, blocked in the production of the natural starter [48]. Similarly, feed-ing of sulfur-containing analogs of the precursor pipecolic acid to a rapL mutant blocked in pipecolic acid biosynthe-sis generated a number of sulfide- or sulfoxide-containing rapamycin analogs [65]. None of these naturally produced rapalogs were advanced to clinical trials, however.

Two important scientific and technological develop-ments, briefly reviewed here, took place during this 30 year period that had significant impact on NP discovery: the development of genetic tools for streptomycetes, and the detailed understanding of NP biosynthesis.

Genetic tools for streptomycetes

The development of recombinant DNA (rDNA) technol-ogy in Escherichia coli took place in the early 1970s; NPs played an important role in its development as all vectors used for cloning depended on the use of genes that con-ferred resistance to antibiotics (penicillin, kanamycin, chloramphenicol, etc.) [33]. Development of rDNA tech-nologies in Streptomyces species lagged for about a dec-ade. Some of the initial advancements included the devel-opment of plasmid vectors and protoplast transformation and regeneration techniques for a variety of both academic and industrial strains [e.g., see 27, 76]. Later, bifunctional vectors were constructed that facilitated engineering of secondary metabolite genes in E. coli followed by conju-gation into streptomycetes and site-specific integration into bacteriophage attachment (attB) sites for stable main-tenance of the engineered genes or gene clusters [18, 28]. These advancements were followed by improved genetic engineering methods facilitated by the use of the λ RED recombination system of cloned genes and gene clusters in bacterial artificial chromosomes (BACs) in E. coli, fol-lowed by conjugation and site-specific integration into any of a number of attB sites to facilitate combinatorial bio-synthesis [3, 20, 87, 88]. In addition, all analytical meth-ods developed for determining gene expression (transcrip-tomics, RNA-Seq), enzyme levels (proteomics), precursor supply (metabolomics) in E. coli were directly applicable to streptomycetes.

Biochemistry and genetics of NP biosynthesis Biochemical pathways for the production of penicillins and cephalosporins were determined by the middle 1960s, employing feeding experiments and biochemical assays on isolated enzymes [97]. When DNA cloning in actinomy-cetes and DNA sequencing became available, biochemical pathways of NPs were inferred largely from the sequences of the genes that determined the corresponding enzymes, although enzymology was always prominent in academic labs interested in NP biosynthesis. Involvement of genes in biochemical pathways was often determined by gene disruption, followed in some instances by complementa-tion analysis. As more biosynthetic genes were sequenced, informatics was used to construct the biochemical steps determined by the predicted enzymes. This paradigm has been used for more than 100 NPs where pathways have been verified genetically, as well as for hundreds more of their structural relatives for which genetic verifica-tion is lacking. All of these data established that, overall, biochemical pathways of NPs appear to contain discrete, single function enzymes that act on and release diffusible compounds, and large multifunctional enzyme complexes that carry out many processive enzymatic steps in a linear order where all compounds are tethered to the complex in the form of thioester linkages. A brief overview is pro-vided here. Detailed descriptions of biochemical pathways, genetics of NP biosynthesis, and structural information of multifunctional enzymes can be found in other reviews [54, 56, 71, 111, 128]. Aminoglycosides and other NPs consist-ing exclusively of sugars, deoxysugars, or acylated sugars, are produced by discrete single-function enzymes. The tripeptide backbones of penicillins and cephalosporins, the tetrapeptide subtilisin, and the cyclic tridecapeptide dap-tomycin are each produced by enzyme complexes collec-tively named non-ribosomal peptide synthetases (NRPS). NRPSs consist of modules, which themselves consist of domains that are responsible for the incorporation of one selected amino acid into the nacent polypeptide chain. The peptide backbone of the glycopeptide vancomycin consist-ing of seven amino acids is biosynthesized by three NRPS proteins containing a total of seven modules. The sugar, vancosamine, is generated from glucose by seven discrete enzymes. Interestingly, the NRPS that produces the cyclic decapeptide cyclosporin A consists of a single protein con-taining ten modules. Similarly, the large macrocyclic back-bones of the polyketides erythromycin, tylosin, spinosyn A, avermectin, amphotericin B, etc., are produced by specific multifunctional enzymes designated polyketide synthases (PKSs) which incorporate, in a processive fashion, specific

acyl-CoA precursors, into a growing acyl (polyketide) chain which is tethered to the multienzyme as a thioester until released at the termination of synthesis. These type I PKSs are also composed of modules which themselves consist of enzymatic domains responsible for a single step in the biosynthesis of the polyketide backbone. The sugar components of erythromycin, tylosin, and others are pro-duced by discrete enzymes.

Early cloning experiments, followed by sequencing of biosynthesis genes initially, and entire genomes afterward consistently revealed that the genes for entire pathways of NP biosynthesis were clustered, even in eukaryotes (e.g., fungi) whose genomes are composed of multiple chromo-somes. The erythromycin biosynthesis cluster consists of three large PKS genes (35 kb) flanked by 17 genes involved in the biosynthesis of sugars, hydroxylation of the pol-yketide backbone, methylatation of one of the sugars, and a thioesterase, as well as a gene conferring resistance to erythromycin [113]. The entire cluster is greater than 50 kb. Some NP biosynthesis clusters are much larger. Some clus-ters (e.g., tylosin [34]), contain a number of positive and negative regulatory genes. Clustering facilitates the clon-ing of the entire NP biosynthesis pathway into vectors, as described above, for production of NPs in heterologous hosts, or for combinatorial biosynthesis.

Combinatorial biosynthesis

At Eli Lilly and Company, advances in genetic engineering methodologies facilitated the generation of novel polyke-tides and glycopeptides. Building on the precursor-directed biosynthesis studies described earlier [7], scientists at Lilly cloned the 4″-mycarosyl isovaleryl-CoA transferase gene (carE) from Streptomyces thermotolerans and expressed it in Streptomyces ambofaciens, the spiramycin producer [44]. Expression of carE in S. ambofaciens caused the pro-duction of isovalerylspiramycin. This served as an early validation of what would later be termed combinatorial biosynthesis [10]. A second example was the reprogram-ming of the polyketide synthase involved in spiramycin biosynthesis by exchanging a module with a different acyl-transferase specificity to produce a novel hybrid polyketide [72]. A novel glycopeptide was generated by cloning the glucosyltransferase gene (gtfE) from the vancomycin pro-ducer, A. orientalis, into the A47934 producer, Streptomy-ces toyocaensis [107]. This was the first novel glycopeptide produced by genetic engineering, and it further validated the concept of combinatorial biosynthesis in Streptomyces species.

At Cubist Pharmaceuticals, the use of λ RED recom-bineering of lipopeptides cloned in BAC vectors facilitated combinatorial biosynthesis around the core cyclic peptide structures of daptomycin and A54145 to generate about 120 novel derivatives for evaluation as potent antibacterial agents active against Gram-positive pathogens [3, 4, 20, 87, 88]. The work at Cubist helped define the “rules” for suc-cessful engineering of NRPS pathways, and demonstrated that complex pharmacological properties can be modulated by changing the primary amino acid sequences of the tride-capeptides of daptomycin and A54145 in ways not address-able by synthetic chemistry.

Combinatorial biosynthesis of modular PKSs has had limited successes at Lilly, Abbott, Kosan, and Biotica. Changes to the structure of erythromycin, tylosin, pikromy-cin, epothilone and other polyketides were initially accom-plished by domain swaps employing cumbersome and time consuming double reciprocal, two step recombination in the producing organism [93, 100, 116, 117]. At Kosan, the erythromycin PKS was re-synthesized employing E. coli codon usage and unique restriction sites so that domain swaps could be done combinatorially using restriction site cloning leading to multiple changes in the structure of the erythromycin polyketide backbone [77]. In addition, scien-tists at Kosan showed that the terminal two modules of the erythromycin PKS, carried on a single PKS polypeptide, could be replaced by the last two modules of the pikro-mycin or oleandomycin PKSs to yield hybrid polyketides. This work was carried out by combining plasmids carrying separate PKS modules from the ery, pik, or ole PKSs in the host Streptomyces lividans [116]. At Kosan, hybrid PKS composed from the tylosin and chalcomycin PKSs or the tylosin and spiramycin PKSs produced the expected hybrid macrolactone backbones. When these hybrid PKSs were placed in a strain of Streptomyces fradiae deleted for the native PKS genes, the hybrid macrolactones produced were then acted upon by the tylosin tailoring enzymes (glycosyl transferases, hydroxylases, methytransferases) to produce novel, fully elaborated NPs [99]. More progress in PKS engineering has been made in recent years as better under-standing of PKS structure has emerged, but the promise of new polyketide-based drugs has not yet been realized. Genomics?based approaches (2000s and beyond)

In the early 2000s, the first two Streptomyces genome sequences revealed the striking observation that many more secondary metabolite gene clusters (SMGCs) are encoded in these large genomes than predicted from their expressed secondary metabolomes [24, 31, 58, 59]. These observations have since been generalized to other actino-mycetes with large genomes [6, 14, 17, 23, 57, 62, 84, 90]. It is now estimated that <10 % of SMGCs are expressed in sufficient quantities to be observed under routine fermenta-tion analyses, and the others require special conditions or genetic manipulations to reveal their products [21, 89, 133,

134]. Among the actinomycetes, the number of SMGCs varies widely [14, 84], and actinomycetes with very large genomes tend to be the most “gifted” [17, 19]. An extreme example is Streptomyces rapamycinicus (a rapamycin pro-ducer) which has a 12.7 Mb genome with 3.0 Mb (24 %) devoted to the biosynthesis of 48 SMGCs [23]. Sequence information also revealed the high frequencies of biosyn-thetic clusters of the NPs discovered in the early discovery period, as well as the presence of antibiotic “resistomes” (multiple genes for antibiotic resistance) among soil iso-lates [131]. Indeed, current knowledge on mechanisms of antibiotic resistance can be exploited to select directly for producers of vancomycin and related glycopeptides from soil actinomycetes [119]. This concept can be extended to other antibiotic classes where the mechanism of antibacte-rial resistance is known and easily selectable.

With inexpensive genome sequencing, advances in understanding secondary metabolite biosynthesis, and advances in analytical and bioinformatics methods, it is now possible to predict at least partial structures of new SMs related to known SMs, and truly novel SMs by scan-ning DNA sequences [78, 120, 126]. Although gene clus-ter sequences are not yet available for all known SMs, this process continues to improve as more SMGC sequences become available. Predicting new and novel pathways from genome sequence data bypasses the tedious task of derep-lication of known compounds, which has been the bane of natural products discovery in recent years [9, 11, 13]. Fur-thermore, improved technology for DNA synthesis, clon-ing, and heterologous expression allows the rapid trans-plantation of novel NP clusters from the sequenced host into well characterized, genetically tractable engineered hosts that have high levels of needed precursors, and con-stitutively expressed strong promoters, together with new high-throughput MS detection methodology [105, 132], to enable high probability production and detection of novel NPs [21].

Genome mining and activation of cryptic pathways

As described above, it is now apparent that actinomy-cetes with large genomes encode multiple (mostly cryptic) SMGCs [1, 6, 23, 24, 31, 58, 59, 90] ranging from ~20 to 50 per genome. It is now feasible to sequence 10,000 [62] to over 100,000 actinomycete genomes in pools (Warp Drive Bio, unpublished data) to identify new and novel SMGCs. There are a number of genetic and fermentation methods to activate the expression of cryptic or poorly expressed SMGCs. Genetic methods include: (1) chemical and transposon mutagenesis; (2) gene cluster duplication/ amplification; (3) overexpression of positive regulators;

(4) disruption of negative regulators; (5) modulation of the transcription apparatus; (6) modulation of the translation apparatus; (7) modulation of post translation of carrier proteins; (8) heterologous expression; (9) synthetic biol-ogy refactoring of promoters and ribosome binding sites; and (10) combinations of (1) through (9). These and other methods to activate or improve the production of SMs are reviewed in detail elsewhere [15, 16, 21].

Aside from exploring multiple fermentation media, there are a number of elicitors or stimulators of SM production, including certain types of stress or chemicals that can be added to fermentations (e.g., heat, ethanol, rare earth ele-ments, glucosamine, and others) [89, 133, 134] or antibiot-ics added at sub-inhibitory concentrations [5, 115]. This is an area ripe for future investigations.

Advances in combinatorial biosynthesis

Early attempts at combinatorial biosynthesis as a means to produce novel NP structures were limited by a lack of understanding of the fundamental structural requirements of these megasynthases for activity. Much progress has been made in understanding the important protein–protein interactions critical to the functioning of PKS and NRPS megaenzymes [12, 20, 40, 66, 73, 128, 130]. In addition, much is now known about effective approaches for com-binatorial biosynthesis of post-PKS and other modifica-tions of SMs [12, 51, 91, 92]. Combinatorial biosynthesis can also be coupled with medicinal chemistry to further expand the numbers and variety of compounds available for evaluation [67]. With the rapid accumulation of new DNA sequences for novel PKS, NRPS and other SMGCs afforded by inexpensive microbial genome sequencing, which can serve as important new sources of “parts and devices” for combinatorial biosynthesis, it is time to rein-vigorate this important approach as an adjunct to microbial genome mining for drug discovery.

The future of natural product discovery

We believe that three key elements will drive natural prod-ucts discovery from microbes in the coming years. 1. We have already learned that microbes (particularly actino-mycetes) with large genomes encode multiple SMGCs, most of which are cryptic, and many of which appear to encode novel SMs. Many more cryptic pathways will be uncovered by inexpensive genome sequencing. Though structural details of truly novel SMs cannot be predicted accurately by bioinformatics alone, the current programs are rapidly improving their predictive power and can read-ily identify truly novel SMGCs [61, 78, 105, 120, 126]. These novel gene clusters can be expressed by genetic and physiological manipulations of the producing microbes or by expression in a heterologous hosts. 2. Scientists have

sampled a miniscule fraction of soil environments [9, 13] and metagenomic and other studies indicate that there exists a wealth of novel SMGCs yet to be discovered [32, 64, 79, 94, 106]. Vastly expanded sampling of soil will provide an inexhaustible source of new microbes (cultiva-ble or not) for genome mining. 3. Advances in structural biology of complex PKS megaenzymes [40, 128, 130] and in functional analysis of engineered NRPS megaenzymes [20] bode well for future applications of combinatorial biosynthesis to exploit novel PKS and NRPS modules and domains identified by genome mining. These three cor-nerstones will be supported by continuing advancements in enzymology, structural biology, bioinformatics, tran-scriptomics, proteomics, metabolomics, and advances in mass spectral analysis. In addition, as understanding of cellular function and the molecular basis of human disease increases, more “smart-screens” will be developed to find NPs with desired activities. Indeed, we predict a second “Golden Age for Antibiotics” and natural product discov-ery. Stay tuned!

References

1. Aigle B, Lautru S, Spiteller D, Dickschat JS, Challis GL, Leb-

lond P, Pernodet JL (2014) Genome mining of Streptomyces ambofaciens. J Ind Microbiol Biotechnol 41:251–264

2. Alberts AW, Chen J, Kuron G et al (1980) Mevinolin: a highly

potent competitive inhibitor of hydroxymethylglutaryl-CoA reductase and a cholesterol-lowering agent. Proc Natl Acad Sci USA 77:3957–3961

3. Alexander D, Rock J, He X, Miao V, Brian P, Baltz RH (2010)

Development of a genetic system for lipopeptide combinatorial biosynthesis in Streptomyces fradiae and heterologous expres-

sion of the A54145 biosynthetic gene cluster. Appl Environ Microbiol 76:6877–6887

4. Alexander D, Rock J, Gu JQ, Mascio C, Chu M, Brian P, Baltz

RH (2011) Production of novel lipopeptide antibiotics related to A54145 by Streptomyces fradiae mutants blocked in biosyn-

thesis of modified amino acids and assignment of lptJ, lptK and lptL gene functions. J Antibiot 64:79–87

5. Amando SI, Sakurai T, Endo K et al (2011) A cryptic antibiotic

triggered by monensin. J Antibiot 64:703

6. Bachmann BO, Van Lanen SG, Baltz RH (2014) Microbial

genome mining for accelerated natural products discovery: is a renaissance in the making? J Ind Microbiol Biotechnol 41:175–184

7. Baltz RH (1982) Genetics and biochemistry of tylosin produc-

tion: a model for genetic engineering in antibiotic-producing Streptomyces. Basic Life Sci 19:431–444

8. Baltz RH (2005) Natural product discovery and development at

Eli Lilly and Company: one scientists view. SIM News 55:5–16 9. Baltz RH (2005) Antibiotic discovery from actinomycetes:

will a renaissance follow the decline and fall? SIM News 55:186–196

10. Baltz RH (2006) Combinatorial biosynthesis of novel antibi-

otics and other secondary metabolites in actinomycetes. SIM News 56:148–160 11. Baltz RH (2006) Marcel Faber Roundtable: is our antibiotic

pipeline unproductive because of starvation, constipation or lack of inspiration? J Ind Microbiol Biotechnol 33:507–513 12. Baltz RH (2006) Molecular engineering approaches to peptide,

polyketide and other antibiotics. Nat Biotechnol 24:1533–1540 13. Baltz RH (2007) Antimicrobials from actinomycetes: back to

the future. Microbe 2:125–131

14. Baltz RH (2008) Renaissance in antibacterial discovery from

actinomycetes. Curr Opin Pharmacol 8:557–563

15. Baltz RH (2010) Streptomyces and Saccharopolyspora hosts for

heterologous expression of secondary metabolite gene clusters.

J Ind Microbiol Biotechnol 37:759–772

16. Baltz RH (2011) Strain improvement in actinomycetes in the

postgenomic era. J Ind Microbiol Biotechnol 38:657–666

17. Baltz RH (2011) Function of MbtH homologs in nonribosomal

peptide biosynthesis and applications in secondary metabolite discovery. J Ind Microbiol Biotechnol 38:1747–1760

18. Baltz RH (2012) Streptomyces temperate bacteriophage integra-

tion systems for stable genetic engineering of actinomycetes (and other organisms). J Ind Microbiol Biotechnol 39:661–672 19. Baltz RH (2014) MbtH homology codes to identify gifted

microbes for genome mining. J Ind Microbiol Biotechnol 41:357–369

20. Baltz RH (2014) Combinatorial biosynthesis of cyclic lipopep-

tide antibiotics: a model for synthetic biology to accelerate the evolution of secondary metabolite biosynthetic pathways. ACS Synth Biol 3:748–759

21. Baltz RH (2015) Genetic manipulation of secondary metabo-

lite biosynthesis for improved production in Streptomyces and other actinomycetes. J Ind Microbiol Biotechnol. doi:10.1007/

s10295-015-1682-x

22. Baltz RH, Seno ET (1981) Properties of Streptomyces fradiae

mutants blocked in biosynthesis of the macrolide antibiotic tylosin. Antimicrob Agents Chemother 20:214–225

23. Baranasic D, Gacesa R, Starcevic A et al (2013) Draft genome

sequence of Streptomyces rapamycinicus strain NRRL 5491, the producer of the immunosuppressant rapamycin. Genome Announc 1:e00581–e00613

24. Bentley SD, Chater KF, Cerde?o-Tárraga AM et al (2002)

Complete genome sequence of the model actinomycete Strepto-

myces coelicolor A3(2). Nature 417:141–147

25. Bérdy J (2012) Thoughts and facts about antibiotics: where we

are now and where we are heading. J Antibiot 65:385–395

26. Bhanot A, Sharma R, Noolvi MN (2011) Natural sources as

potential anti-cancer agents: a review. Int J Phytomed 3:9–26

27. Bibb MJ, Ward JM, Hopwood DA (1978) Transformation of

plasmid DNA into Streptomyces at high frequency. Nature 274:398–400

28. Bierman M, Logan R, O’Brien K, Seno ET, Rao RN, Schoner

BE (1992) Plasmid cloning vectors for the conjugal trans-

fer of DNA from Escherichia coli to Streptomyces spp. Gene 116:43–49

29. Breton RC, Reynolds WF (2013) Using NMR to identify and

characterize natural products. Nat Prod Rep 30:501–534

30. Butler MS, Robertson AA, Cooper MA (2014) Natural product

and natural product derived drugs in clinical trials. Nat Prod Rep 31:1612–1661

31. Challis GL (2014) Exploitation of the Streptomyces coelicolor

A3(2) genome sequence for discovery of new natural prod-

ucts and biosynthetic pathways. J Ind Microbiol Biotechnol 41:219–232

32. Charlop-Powers Z, Owen JG, Reddy BVB, Ternei MA, Guima-

raes DO, de Frias UA, Pupo MT, Seepe P, Feng Z, Brady SF (2015) Global biogeographic sampling of bacterial secondary metabolism. eLife 4:e05048

33. Cohen SN, Chang AC, Boyer HW, Helling RB (1973) Con-

struction of biologically functional bacterial plasmids in vitro.

Proc Natl Acad Sci USA 70:3240–3244

34. Cundliffe E (2008) Control of tylosin biosynthesis in Strepto-

myces fradiae. J Microbiol Biotechnol 18:1485–1491

35. Debono M, Willard KE, Kirst HA et al (1989) Synthesis and

antimicrobial evaluation of 20-deoxo-20-(3,5-dimethylpiperi-

din-1-yl)desmycosin and related cyclic amino derivatives. J Antibiot 42:1253–1267

36. Demain AL (2014) Importance of microbial natural products

and the need to revitalize their discovery. J Ind Microbiol Bio-

technol 41:185–201

37. Denizot F, Lang R (1986) Rapid colorimetric assay for cell

growth and survival. Modifications to the tetrazolium dye pro-

cedure giving improved sensitivity and reliability. J Immunol Methods 89:271–277

38. Denoya CD, Fedechko RW, Hafner EW et al (1995) A second

branched-chain α-keto acid dehydrogenase gene cluster (bkd-

FGH) from Streptomyces avermitilis: its relationship to aver-

mectin biosynthesis and the construction of a bkdF mutant suitable for the production of novel antiparasitic avermectins. J Bacteriol 177:3504–3511

39. DiMarco A, Gaetani M, Dorigotti L, Soldati M, Bellini O

(1964) Daunomycin: a new antibiotic with anti-tumor activity.

Cancer Chemother Rep 38:31–38

40. Dutta S, Whicher JR, Hansen DA et al (2014) Structure of a

modular polyketide synthase. Nature 510:512–517

41. Eisenstein BI, Oleson FB Jr, Baltz RH (2010) Daptomycin:

from the mountain to the clinic with the essential help from Francis Tally, MD. Clin Inf Dis 50:S10–S15

42. Endo AMK (1979) A new hypo-cholesteremic agent produced

by a Monascus species. J Antibiot 32:852–854

43. Endo A (2010) A historical perspective on the discovery of the

statins. Proc Jpn Acad Ser B 86:484–493

44. Epp JK, Huber ML, Turner JR, Goodson T, Schoner BE (1989)

Production of a hybrid macrolide antibiotic in Streptomyces ambofaciens and Streptomyces lividans by introduction of a cloned carbomycin biosynthetic gene from Streptomyces ther-

motolerans. Gene 85:293–301

45. Galm U, Sparks TC (2015) Natural product derived insecti-

cides: discovery and development of spinetoram. J Ind Micro-

biol Biotechnol. doi:10.1007/s10295-015-1710-x

46. Gallo RC, Whang-Peng J, Adamson RH (1971) Studies on the

antitumor activity, mechanism of action, and cell cycle effects of camptothecin. J Nat Cancer Inst 46:789–795

47. Giddings LA, Newman DJH (2013) Microbial natural products:

molecular blueprints for antitumor drugs. J Ind Microbiol Bio-

technol 40:1181–1210

48. Gregory MA, Petkovic H, Lill RE, Moss SJ, Wilkinson B, Gais-

ser S, Leadlay PF, Sheridan RE (2005) Mutasynthesis of rapa-

mycin analogues through the manipulation of a gene governing starter unit biosynthesis. Angew Chem Int Ed 44:4757–4760

49. Havlicek V, Lemr K, Schug KA (2013) Current trends in

microbial diagnostics based on mass spectrometry. Anal Chem 85:790–797

50. Heeb S, Fletcher MP, Chhabra SR, Diggle SP, Williams P,

Cámara M (2011) Quinolones: from antibiotics to autoinducers.

FEMS Microbiol Rev 35:247–274

51. Heide L (2014) New aminocoumarin antibiotics as gyrase

inhibitors. Int J Med Microbiol 304:31–36

52. Henderson DJ, Naya I, Bundick RV, Smith GM, Schmidt JA

(1991) Comparison of the effects of FK-506, cyclosporine A and rapamycin on IL-2 production. Immunology 73:316–321 53. Hendlin D, Stapley EO, Jackson M et al (1969) Phosphonomy-

cin, a new antibiotic produced by strains of Streptomyces. Sci-

ence 166:122–123

54. Hertweck C (2015) Decoding and reprogramming complex pol-

yketide assembly lines: prospects for synthetic biology. Trends

Biochem Sci 40:189–199

55. Heusler K, Pletscher A (2001) The controversial early history of

cyclosporine. Swiss Med Wkly 131:299–302

56. Hur GH, Vickery CR, Burkart MD (2012) Explorations of cata-

lytic domains in non-ribosomal peptide synthetase enzymology.

Nat Prod Rep 29:1074–1098

57. Iftime D, Kylik A, H?rtner T et al (2015) Identification and acti-

vation of novel biosynthetic gene clusters by genome mining in

the kirromycin producer Tü 365. J Ind Microbiol Biotechnol.

doi:10.1007/s100295-015-1685-7

58. Ikeda H, Ishikawas J, Hanamoto A, Shinose M, Kikuchi H,

Shiba T, Sakaki Y, Hattori M, ōmura S (2003) Complete genome sequence of and comparative analysis of the indus-

trial microorganism Streptomyces avermitilis. Nat Biotechnol 21:526–531

59. Ikeda H, Shin-ya K, Omura S (2014) Genome mining of the

Streptomyces avermitilis genome and development of genome-

minimized hosts for heterologous expression of biosynthetic gene clusters. J Ind Microbiol Biotechnol 41:233–250

60. Itoh T, Ishii K, Irikura T, Ueno Y, Kojima A, Horie Y (1993) A

modified method of mixed lymphocyte reaction: establishment

of the assay system and its application to extracts of fungal cul-

tures. J Antibiot 46:1575–1581

61. Johnston CW, Connaty AD, Skinnider MA, Li Y, Grunwald

A, Wyatt MA, Kerr RG, Magarvey NA (2015) Informatic search strategies to discover analogues and variants of natural

product archetypes. J Ind Microbiol Biotechnol. doi:10.1007/

s10295-015-1675-9

62. Ju KS, Gao J, Doroghazi JR et al (2015) Discovery of

phosphonic acid natural products by mining the genomes of 10,000 actinomycetes. Proc Natl Acad Sci USA 112:12175–12180

63. Kanomori H, Shima T, Morita C, Hata T (1957) Studies on the

antitumor activity of mitomycin. J Antibiot 10:120–127

64. Katz M, Hover BM, Brady SF (2015) Culture-independent dis-

covery of natural products from soil metagenomes. J Ind Micro-

biol Biotechnol. doi:10.1007/s10295-015-1706-6

65. Khaw LE, B?hm GA, Metcalf S, Staunton J, Leadlay PF (1998)

Mutational biosynthesis of rapamycins by a strain of Streptomy-

ces hygroscopicus NRRL 5491 disrupted in rapL, encoding a

putative lysine cyclodeaminase. J Bacteriol 180:89–814

66. Khosla C, Hershlag D, Cane DE, Walsh CT (2014) Assembly

line polyketide synthases: mechanistic insights and unsolved problems. Biochemistry 53:2875–2883

67. Kim E, Moore BS, Yoon YJ (2015) Reinvigorating natural

product combinatorial biosynthesis with synthetic biology. Nat

Chem Biol 11:649–659

68. Kinch MS, Haynesworth A, Kinch SL, Hoyer D (2014) An

overview of FDA-approved new molecular entities: 1827–2013.

Drug Discov Today 19:1033–1039

69. Kirst HA (2010) The spinosyn family of insecticides: real-

izing the potential of natural products research. J Antibiot 63:101–111

70. Koehn FE, Carter GT (2005) The evolving role of natural prod-

ucts in drug discovery. Nat Rev Drug Discov 4:206–220

71. Kudo F, Eguchi T (2009) Biosynthesis genes for aminoglyco-

side antibiotics. J Antibiot 62:471–481

72. Kustoss S, Huber M, Turner JR, Paschal JW, Rao RN (1996)

Production of a novel polyketide through the construction of a

hybrid polyketide synthase. Gene 183:231–236

73. Ladner CC, Williams GJ (2015) Harnessing natural product

assembly lines: structure, promiscuity, and engineering. J Ind

Microbiol Biotechnol. doi:10.1007/s10295-015-1704-8

史上最全的外语类科研项目申报干货汇总.

Part One 宏观指导 彭青龙 上海交通大学外国语学院教授、副院长、博士生导师 研究领域:外语教学方法、专门用途英语、其他外国文学研究、其他外国社会文化 主持项目:独立主持课题十项,其中国家社科基金重大课题子项目一项、国家社科基金一般项目两项、教育部人文社科基金一般项目一项、澳大利亚政府基金项目两项等。 Q:如何更好地利用国家社科基金项目、教育部人文社会科学研究项目中的选题指南来进行选题? a.课题申报者应仔细研究课题指南,深刻领会其内涵和外延,确定适合自己的选题。国家项目课题指南往往反映的是最前沿或者最基础却尚未解决好的研究问题。 b.国家项目课题指南每年都会有变化,课题申报者可以对比研究近两三年课题指南的变化,较往年有变化的地方通常是国家特别希望在这个领域内进行着力研究的问题。 Q:如何判断某一个课题是否具有学术价值或应用价值?可否举例说明? 一个课题的学术价值体现在其理论价值和应用价值两方面。而课题的创新往往体现在观点创新或是方法创新两个主要方面。如,某课题一般运用定性研究的方法,而本人综合运用了定性和定量的两种方法,这从某种意义上来说就是一种创新,方法创新也可理解为新路径或新视角。创新往往是站在巨人的肩膀上的创新。Q:国家课题的选题与一般学术论文选题区别在哪里? 国家课题往往是各领域亟待解决的问题。研究成果受众面大,具有广泛的应用价值或理论价值,也具有相当的示范性。国家课题一般是集中某一重要方面进行多维度的探讨,更具系统性和逻辑性,其成果至少应该是系列论文而非单篇文章。Q:课题申报书常见问题有哪些? 申报书需要展示的是三个方面,即why, what, how, 即你为什么要做此项研究,研究的主要内容是什么,你将如何组织实施你的研究。同时,前期积累也很重要。总之,通过申报书,你要能说服评审专家可以将这个项目放心地交给你。 课题申报书中的常见问题有: a. 选题本身存在概念不清、逻辑混乱。申报者对基本概念应有明晰的判断,主概念和子概念之间关系要有逻辑性。 b. 选题过大或过小。有些选题太大,可写成大型系列丛书,但申报书中的论述不够详实,难以支撑。有些选题也很大,但仅以很小的案例做支撑,论证充分性不够,难以令人信服。 c. 文献综述,述而不论。一些申报书中文献综述仅罗列多方观点,却没有批判性的论述,或将自己的论述淹没在描述中,没有鲜明地提出自己的观点。文献综述在综合分析前人研究成果的同时,要对其进行评价和判断,即哪些观点是得到普遍认同的,哪些观点还存在不足,有待进一步研究。而本课题研究正是弥补不

地震处理及解释软件发展现状

地震处理及解释软件发展现状 作者:发布时间:2010-04-08 10:51:27 地震资料处理技术的发展与计算机技术的发展息息相关。从模拟处理到数字处理;从简单的陆上二维资料处理到复杂的山地资料处理、全三维资料处理、高分辨率和深层资料处理等;从常规资料的处理到处理解释一体化的叠前深度偏移技术,每一次地球物理技术的进步都离不开计算机技术的进步和应用软件的发展。 以胜利油田的地震资料处理计算机装备为例,其发展过程已历经了数代的变化。从最早的IRIS60机、TIMAP—I、TIMAP4、VAX11/782、IBM3083,到并行计算SGI/Orgin2000和IBM—SP,以及目前正在迅猛发展的PC—CLUSTER,运算速度已从最初的每秒40万次提高到现在的每秒万亿次。 随着地震资料处理硬件装备的发展,处理软件也在不断地更新,处理技术日趋完善。勘探软件是现代地震勘探和油藏描述的基本必备工具,自上世纪70年代,国外的一些软件公司就已着手开发地震处理及解释软件系统,并初步形成了商业化软件,开始在全世界范围内推广和应用。进入上世纪90年代,比较成熟的处理软件有西方地球物理公司的Omega处理软件、法国CGG公司的GEOVECTEUR PLUS处理软件、LandMark公司的Promax处理软件、帕拉代姆公司的GeoDepth软件、Focus软件。国内较早从事勘探软件研究和开发的单位,主要是以东方地球物理公司(原石油物探局)为主,它的处理软件为Grisys处理软件。这些软件的处理技术水平各具特色。另外,随着油藏地球物理技术的发展,各种相关的特殊处理软件逐步发展与完善。 地震数据处理软件的发展 批处理阶段上世纪70~80年代末,由于计算机技术落后,限制了地震处理软件和处理技术的发展,地震处理软件一直处于批处理阶段,代表性的软件有:法国CGG公司的GEO—MASTER软件、美国GSI公司的TIPEX软件、美国WGC公司的IQ处理软件、美国CSD 公司DISCO软件等。 交互处理阶段上世纪90年代初,随着计算机技术的飞速发展,地震处理软件和处理技术发展很快。开始发展交互地震处理软件。代表性的软件有:法国CGG公司的

广东省哲学社会科学“十三五”规划项目外语专项申请书【模板】

广东省哲学社会科学“十三五”规划项目 外语专项 申请书 项目类别 项目名称 项目负责人 负责人所在单位 填表日期

广东省哲学社会科学规划领导小组办公室制 二О二О年六月 申请者的承诺: 保证如实填写本表各项内容。如果获准立项,承诺以本表为有约束力协议,遵守《广东省哲学社会科学规划项目管理办法》等有关规定,认真开展研究工作,取得预期研究成果。 申请者(签章): 年月日 填表注意事项 一、本表请如实填写。 二、本表第一项“项目负责人、主要参加者情况”部分栏目填写说明: 1、研究类型:指本项目研究属基础理论研究、应用开发研究、综合研究等。 2、主要参加者:必须真正参加本项目研究工作,不含项目负责人,不包括科研、财务管理人员。 3、预期成果形式:指最终成果形式,含专著、研究报告、论文等。其中,论文指内容具有相关性、系统性的,已发表及未发表的论文若干篇。 4、项目完成时间:1-2年。 三、申请人具有副高以上(含副高)职称或具有博士学位者,不填“推荐人意见”。 四、本表内“申请者(签章)”和“推荐人姓名”处须手写,不能打印。

五、本表用A3纸双面印制,中缝装订成册。 六、本表由项目负责人所在单位加具单位意见,并统一报送省哲学社会科学规划领导小组办公室。 七、省哲学社会科学规划领导小组办公室地址:XX市天河北路618号广东社科中心B座9楼,邮编:******,电话:(020)********、********。 一、项目负责人、主要参加者情况

二、课题设计论证

三、项目负责人正在主持的其他项目 四、推荐人意见 五、项目负责人所在单位意见

地震解释的现状及发展趋势

地震波地质信息综合解释 摘要:地震解释质量决定了一个区块勘探开发的方向和进程,地震解释的发展对解释人员提出了更高的要求,即要求解释人员通晓地质知识,同时具有物探知识。本文主要从现今已经在应用的解释技术和方法以及近年来涌现出来的一些新思路、新方法展开论述。分别包括三维可视化技术、构造解释、构造解释和利用振幅属性预测含烃概率、利用波峰瞬时频率计算薄层厚度、多子波地震道分解和重构等。 关键字:地震解释、构造解释、振幅属性、波峰瞬时频率 引言:地震资料解释是勘探和开发地震的最后环节,其功能是将地震信息翻译成地质语言或符号;其目的是直接服务于勘探和开发。因此解释质量决定了一个区块勘探开发的方向和进程。地震勘探开发技术发展的目标都是为了提供更好的易于解释的具更高可信度的地震资料。地震解释现在更多地强调综合性和在地质规律控制下的地震解释。这对解释人员提出了更高的要求,即要求解释人员通晓地质知识,同时具有物探知识。地震解释从来就不是从事物探方法研究人员单纯可以从事的工作。地震解释已经开始从注重地震解释方法向注重多学科综合性的转变,现在更为明显!地震解释的另一个明显的趋势是强调在地质规律认识下的地震解释,即地震和地质的紧密结合。 一、地震综合解释的现今技术及方法 在地震综合解释方面,主要是以地震反演技术、多种属性分析技术及三维解释为主体的地震综合储层预测技术,通过与层序地层学、测井和地质等其他测量解释成果的结合给出地震资料综合解释的应用实例。例如AmoutColpaert应用神经网络将地震解释数据和井中岩石物理特性分析联合实现多属性分析,从而进行岩相预测。靶区的目标地层是岩溶发育的斜坡形向陆架坡过渡的碳酸盐岩地层,探区内井资料很少或几乎没有,作者综合应用了基于井资料的层序地层分析、岩石物理分析和多属性地震分析,对无井控制区的岩相进行了预测。其基本流程见图1。

英语课题立项申报书详解

伊川县江左镇中基 础教育教研课题 立项申报书 学 科 分 类_____初中英语______________________ 课 题 名 称_多媒体课件优化中学生英语阅读的实践研究 课 题 主 持 人___ 刘志刚___________________________ 课 题 组 成 员_韩世伟 程会英 黄爱香 杨玉温 端木梦梦 主持人工作单位____ 伊川县江左镇初级中学_________ 申 请 日 期__ 2014年12月10日 伊川县教育局基础教育教研室 立项编号 ?yckt160803 学科代码 08?

填表说明 一、本表须经课题主持人所在单位和中心校审核,签署明确意见,承担信誉保证并加盖公章后,方可上报。 二、封面左上方代码框申请人不填,其他栏目由申请人用中文填写。每项课题主持人限1人;主要参与者不包括课题主持人,至少2人,最多5人。 三、本表报送一式3份,请用A4纸双面打印、复印,于左侧装订成册。同时,须提供本表的电子版1份。 四、请用钢笔或电脑打印,准确如实填写各项内容,书写要清晰、工整。 五、伊川县教育局基础教育教研室课题管理办公室: 联系人:赵康卷电话:

?一、基本情况 ? 二、 课题 设计论证 课题名称 多媒体课件优化中学生英语阅读的实践研究? 主持人 姓 名 ?刘志刚 政治面貌 党员? 性别 男? 年龄 41 行政职务 教导主任 专业 职称 中一? 学科 专业 英语? 学历 学位 本科 起止时间 2014年 12 月 10 日至 2015 年 12 月 10日 工作单位 通讯地址 伊川县江左镇中 邮政 编码 471314? 固定电话 E-mail 移动电话 主 要 参 与 者 姓 名 性别 年龄 专业 职称 学科 专业 学历学位 工作单位 韩世伟 男? 48 中二? 初中英语 本科 伊川县江左镇中 程会英 女? 40 中二? 初中英语 专科 伊川县江左镇中 黄爱香 女? 37 中二? 初中英语 本科 江左教育? 杨玉温? 女? 41 中二? 初中英语 本科 伊川县电力中学 端木梦梦 女? 26 中二? 初中英语 本科 伊川县江左镇中 预 期 成 果 (在选项上打“√” 或加粗) A .专着 B.研究报告 C.论文 D.其他

地震数据处理解释技术发展研究

地震数据处理解释技术发展研究 地震数据处理解释是地震勘探的主要组成部分,是石油天然气勘探开发产业链中对油田勘探开发效益影响最大、技术含量最高的一环。…… 一、地震数据处理解释是地震勘探的主要组成部分 地震勘探就是通过人工地震反射波“给地球做CT”,让油气勘探者能够“看见”地层的地质构造和油藏情况,为石油公司“找油”做出含油气评价、提出钻井位置、模拟油藏未来的生产动态以便为后续油气藏开采和开发提供技术资料。 地震勘探包括地震采集、处理和解释三大部分:地震采集是利用野外地震采集系统获取地震数据处理所需的反射波数据;地震数据处理的目的是对地震采集数据做各种处理提高反射波数据的信噪比、分辨率和保真度以便于解释;地震解释分为构造解释、地层解释,岩性和烃类检测解释及综合解释,目的是利用地震反射波的地质特征和意义确定井位寻找石油。地震数据处理依赖于地震采集数据的质量,处理结果直接影响解释的正确性和精确度和找油的成功率。 图1 地震勘探产业链构成 地震数据处理解释是地震勘探的主要组成部分,是石油天然气勘探开发产业链中对油田勘探开发效益影响最大、技术含量最高的一环。其原因有四:1、石油勘探地震数据处理解释与井位部署成功率、油田发现、油田采收率、油田增储上产等经济效益直接相关,是寻找油气资源的关键技术; 2、石油勘探技术发展的基础主要体现在地震数据处理环节中地震成像技术的发展;3、地震数据处理解释下游钻井业务等油气开采技术均十分成熟;4、上游地震数据采集依赖于先进的仪器设备,理论简单。综合而言,地震数据处理的质量和地震成像的准确度与清晰度直接决定油气资源的发现的成败和勘探成功率,是影响后期油田生产建设最重要的环节。 BP公司北海油田日产量与地震数据处理解释新技术的关系表明,新技术尤其是地震成像技术的发展和应用对于油田产量的增加影响极大。 图2 石油勘探地震数据处理解释技术对北海油田的产量的影响由此可见,地震数据处理解释是地震勘探的主要组成部分,其发展和技术进步对于解决人类能源供应问题具有十分重要的意义。 二、地震数据处理解释技术发展历程 地震数据处理解释技术中最核心的就是地震成像技术,因此地震数据处理解释技术的发展历程主要依据地震成像技术的发展水平进行划分。 地震数据处理解释最早出现于20世纪20年代初期。随后的40年间由于是对光点记录(1920—1950)和模拟记录(1950—1965)进行处理,在这一阶段地震处理解释技术发展缓慢,也没有可实用的地震成像技术出现。

1+X+证书制度专项研究+2020+年度课题指南

附件1 1+X证书制度专项研究2020年度课题指南 为深入贯彻《国家职业教育改革实施方案》部署,落实《关于在院校实施“学历证书+若干职业技能等级证书”制度试点方案》等文件要求,着力通过专项课题研究、协同创新,为1+X证书制度目标任务实现、重点难点问题解决提供科研与智力支持,特制定本指南。申请人可结合自己的学术专长和研究基础选择申报,指南选题如下。 1.“放管服”改革背景下职业技能等级证书的功能、定位、效力和话语体系研究 主要内容:在“放管服”改革背景下,系统梳理技术技能人才评价制度、评价模式改革脉络,聚焦对技术技能人才评价维度、内容、方法等方面的改革,研究提出职业技能等级证书的功能、定位和效力,明晰职业技能等级标准、职业技能等级证书、复合型技术技能人才、国家资历框架、职业教育学分银行、培训评价组织等概念与内涵,建构1+X证书制度作为中国特色职业教育制度的话语体系和基本语境,为1+X证书制度理论体系的建构提供建议。 研究周期:1年 预期成果:论文3篇(在CSSCI核心库来源期刊发表至少1篇);

专著(或合著)1部;研究报告1篇(不少于5万字)。 2.职业技能等级证书对接职业标准和教学标准的机制研究 主要内容:在技术技能人才培养培训中实行学历证书和职业技能等级证书相结合的理论依据、实践寻证和教育学价值;1+X证书制度与原“双证书”制度对比研究,德国、澳大利亚、新西兰等国家证书制度的比较研究;在职业院校实施“1+X”复合型技术技能人才培养模式研究;研究提出职业技能等级证书对接国际先进标准、对接职业标准、对接院校教学标准的运行机制和政策建议。 研究周期:10个月 预期成果:阶段性研究报告、总报告、论文、专著。论文3篇(在CSSCI核心库来源期刊发表至少1篇);专著(或合著)1部;总研究报告1篇(不少于5万字),阶段性研究报告不少于2篇。 3.职业技能等级证书效力和待遇落实的实施路径研究 主要内容:梳理国外资历框架制度发展脉络,结合国内实际,通过调查分析,厘清实施中存在的主要问题,找准切入点和突破口,研究提出院校内和院校外实施的职业技能等级证书具备同等效力和待遇、行业企业和用人单位切实兑现相关待遇的具体政策与保障机制,相关学习成果认定、积累和转换等具有同一效能的具体实施路径。 研究周期:10个月

2020年度粤港澳大湾区教育提高专项课题指南

2020年度粤港澳大湾区教育专项课题指南 2020年度粤港澳大湾区教育专项课题分为重大项目和一般项目两种类型。具体如下: 一、重点项目选题 围绕粤港澳大湾区国际教育示范区建设研究 二、一般项目参考选题 1、粤港澳大湾区高校合作办学体制机制研究 2、国际湾区高等教育协同创新发展指数评估研究 3、粤港澳高校学分互认、累计与转换体系研究 4、粤港澳高校科研成果分享转化机制研究 5、粤港澳大湾区特色学院建设研究 6、粤港澳大湾区高校课程开放共享研究 7、粤港澳大湾区大学集群发展研究 8、粤港澳大湾区高校创新创业体系研究 9、粤港澳大湾区高等教育提升国家身份认同的路径与机制 10、粤港澳大湾区高等教育规模、布局与类型优化研究 11、粤港澳大湾区职业教育体制机制创新研究

12、粤港澳大湾区职业教育资历框架研究 13、粤港澳大湾区现代职业教育体系建设研究 14、粤港澳大湾区职业教育1+X证书制度研究 15、粤港澳大湾区现代学徒制研究 16、粤港澳大湾区职业教育园区建设研究 17、粤港澳大湾区基础教育交流合作路径机制研究 18、粤港澳大湾区基础教育与社区教育结合研究 19、粤港澳大湾区基础教育质量标准比较研究 20、粤港澳大湾区学前教育优化发展研究 21、粤港澳大湾区儿童体质与健康促进研究 22、粤港澳大湾区特殊教育融合发展研究 23、广东省港澳子弟学校布局优化及管理机制研究 24、粤港澳大湾区教师资质互认研究 25、粤港澳大湾区家校共育研究 26、随迁子女在粤港澳大湾区平等接受高中教育和参加高考政策研究 27、粤港澳大湾区民办教育发展政策与机制研究 28、粤港澳大湾区教育信息化发展及合作模式研究

初中英语口语课题申报评审书

大连市“十二五”教育科学专项课题申请·评审书 课题类别课程改革研究 课题名称XX地区中小学英语口语教学策略研究 课题负责人张壮 负责人所在单位XXXX实验学校 负责人联系电话1556689XXXX 填表日期2013-5 大连市教育科学规划领导小组办公室印制 2011年5月

申请者的承诺: 我保证如实填写本表各项内容。如果获准立项,我承诺以本表为有约束力的协议,遵守大连市教育科学规划领导小组办公室的有关规定,认真开展研究工作,取得预期研究成果。大连市教育科学规划领导小组办公室有权使用本表所有数据和资料。 申请者(签章):张壮 2013年5月6日 填表说明 1、封面上方2个代码框申请人不填,其它相关栏目由申请人用计算机或钢笔并用中文准确如实填写。本表上报三份,其中“课题论证”部分单独复印5份一并上交。 2、课题类别请按大连市教育科学规划领导小组办公室分步下发的课题指南范围类型填写。如:课程改革研究、德育研究、现代教育技术应用研究、学前教育等。 3、课题名称应准确、简明反映研究内容,最多不超过40个汉字(包括标点符号)。 主题词按研究内容设立。主题词最多不超过3个,主题词之间空一格。 4、课题负责人必须真正参加本课题研究。 主要参与者不含课题负责人,不包括科研管理、财务管理、后勤服务等人员。 负责人所在单位须填写名称加盖公章,表明内容属实,并承担本课题管理任务、信誉保证,提供完成课题的时间、条件。 5、预期成果包括阶段成果和最终成果。其中最终成果形式有:专著、译著、研究报告、工具书、电脑软件及其它。最终成果请选项填写,最多选报2项。 6、经费预算

一、数据表

山东省教育科学“十三五”规划2016—2017年度课题选题指南

附件2 山东省教育科学“十三五”规划2016—2017年度 课题选题指南 一、重大招标课题选题指南 (一)教育科学规划重大招标课题 山东省2030年教育现代化发展指标体系及监测研究 山东省学龄人口变动和学校布局调整预测研究(2016—2025年) 区域义务教育质量监测研究 基于学生发展核心素养的课程体系建构研究 山东省中小学教师专业素养整体提升的体制机制创新研究 山东省推进职业教育集团化办学的支持政策与保障机制研究 山东省高校本科教学质量提升研究 山东省家庭教育指导服务体系建设研究 山东省优秀传统文化教育课程体系构建研究 (二)体育与艺术教育专项重大招标课题 学校体育过程评价体系研究 山东省中小学生艺术素质评价体系构建与实施策略研究 (三)教育招生考试专项重大招标课题 山东省高考改革的考试理论与实践研究 山东省教育考试网上评卷质量监控模型与安全认证技术研究 山东省教育招生考试信息安全与可靠性技术的应用研究 (四)区域教育教学改革重大招标课题 区域推进中小学思维导图课堂教学理论与实践研究 二、专项课题选题指南 (一)传统文化教育专项 优秀传统文化教育与社会主义核心价值观融合研究 优秀传统文化教育师资队伍培养机制和人才库建设研究

优秀传统文化教育与学校教育、家庭教育和社会教育的融合机制研究中小学优秀传统文化教育课程体系建构研究 优秀传统文化教育的学段衔接研究 优秀传统文化教育课程内容的序列化研究 优秀传统文化教育潜在课程开发研究 优秀传统文化与学科课程内容、专业课程内容融合研究 优秀传统文化与中小学语文、思想品德课程内容融合研究 优秀传统文化教育教学组织形式多样化发展研究 优秀传统文化教育案例研究 优秀传统文化教育融入校园文化建设的途径研究 优秀传统文化教育与“互联网+”的融合研究 鲁、台学校优秀传统文化教育比较研究 教师优秀传统文化教育素养提升研究 社会力量参与优秀传统文化教育教学的机制研究 孔子学院总部体验基地运作机制研究 中小学优秀传统文化教育校本课程开发研究 中小学优秀传统文化教育课程整合研究 大学生优秀传统文化教育理论与实践研究 高等学校优秀传统文化教育课程开发研究 优秀传统文化素养评价体系研究 (二)创新创业教育专项 创新创业教育服务区域经济和产业转型升级的机制研究 创新创业人才培养机制研究 创新创业人才培养模式研究 创新创业教育生态系统构建研究 创新创业人才评价体系构建研究 创新创业教育教师能力建设研究 创业学学科建设与发展研究 创新创业教育内容创新研究 创新创业教育与专业教育融合研究 创新创业教育教学方法研究 创新创业教育课程体系构建研究 创新创业教材建设研究

地震勘探技术的发展与应用

地球探测与信息技术 读书报告 课题名称:地震勘探的发展与应用 班级:064091 姓名:吴浩 学号:20091004040 指导老师:胡祥云

地震勘探的发展与应用 吴浩 (地球物理与空间信息学院,地球科学与技术专业) 摘要地震勘探是地球物理勘探中发展最快的一项技术,近年来,高分辨率地震勘探仪器装备、处理软件升级换代速度明显加快,地震资料采集、处理与解释出现了一体化的趋势。从常规的地震勘探发展到二维地震、三维地震、高精度地震勘探等先进技术,应用于石油、煤炭、采空区调查、地热普查等重要领域,由陆地不断向海洋发展。本文着重针对地震勘探过程和技术的发展几个重要阶段及应用进行展开。 关键字地震勘探三维地震石油勘探煤矿发展与应用 1 引言 地震勘探是利用岩石的弹性性质研究地下矿床和解决工程地质,环境地质问题的一种地球物理方法。地震勘探应用领域广泛,与其他物探方法相比,具有精度高、分层详细和探测深度大等优点,近年来,随着电子技术、计算机技术的高速发展,地震勘探的仪器装备、处理软件升级换代的速度明显加快,地震资料采集、处理与解释的一体化趋势得到加强。从常规的地震勘探发展到二维地震、三维地震、高精度地震勘探等先进技术,通常用人工激发地震波,地震波通过不同路径传播后,被布置在井中或地面的地震检波器及专门仪器记录下来,这些地震拨携带有所经过地层的丰富地质信息,计算机对这些地震记录进行处理分析,并用计算机进行解释,便可知道地下不同地层的空间分布,构造形态,岩性特征,直至地层中是否有石油、天然气、煤等,并可解决大坝基础,港口,路,桥的地基,地下潜在的危险区等工程地质问题,以及环境保护,考古等问题。 2 地震勘探过程及发展 地震勘探过程由地震数据采集、数据处理和地震资料解释3个阶段组成。 1.地震数据采集 在野外观测作业中,一般是沿地震测线等间距布置多个检波器来接收地震波信号。常规的观测是沿直线测线进行,所得数据反映测线下方二维平面内的地震信息。一般地讲,地震野外数据采集成本占勘探成本的80%左右,因此世界各国为了降低勘探成本、提高勘探效果,

前沿:海洋宽频带地震勘探新技术扫描

前沿:海洋宽频带地震勘探新技术扫描 文|吴志强 国土资源部海洋油气资源与环境地质重点实验室

1、概况 海洋地震勘探在海洋地质调查、油气藏勘探与开发中起到了无可替代的重要作用。随着勘探领域的不断拓展,地震勘探的难度越来越大。在深部地质调查和复杂构造、火山岩(或碳酸盐岩)屏蔽下的油气藏地震勘探中,为了获取目的层有效反射信号、实现精确成像,对地震数据采集的要求进一步提高,包括采集到低频、高频成分丰富的宽频带、高信噪比原始地震记录。地震信号中的低频信息具有穿透能力强、对深部目的层成像清晰的优势,同时也使地震反演处理结果更具稳定性。宽频带可产生更尖锐子波,为诸如薄层和地层圈闭等重要目标体的高分辨率成像提供全频带基础数据。 理论研究表明:当地震数据的频带宽度不低于两个倍频程时,才能保证获得较高精度的成像效果;频带越宽,地震成像处理的精度越高;增加低频分量的主要作用是减少子波旁瓣,降低地震资料解释的多解性,提高解释成果的精度。 图形象地展示了低频分量的重要性:高频分量丰富、但缺少低频分量的地震子波的主峰尖锐,却会产生子波旁瓣,使地震资料的精确解释变得困难且多解;高分辨率子波是在低频和高频两个方向都得到拓展的宽频带子波,这样子波的主峰尖锐、旁瓣少且能量低,能分辨厚度极小的薄层,地震解释的精度高。 现今地震资料反演处理大多是基于模型的地震反演,成功的关键是能否提取真实子波和建立精确的低频模型。常规地震数据中缺失低频信息,只能采用从测

井数据中提取低频分量再与地震数据反演的相对波阻抗合并处理方式得到绝对 波阻抗。 在目标地质体复杂、钻井少的探区,仅靠测井资料提取的低频分量难以反映复杂地质体横向变化,导致不精确或假的反演结果。为弥补该缺陷,一般采用从地震叠加速度提取低频分量方式,而叠加速度只能提供0~5Hz低频信息,无法弥补常规地震所缺少的0~10Hz低频分量。可见,地震数据中低频信息对保证地震岩性反演的精度意义重大。 然而,在海洋地震勘探中得到宽频带地震数据是比较困难的。 首先,在常规海洋地震数据采集中,电缆和气枪都要以固定深度沉放于海平面之下,以保证下传的激发能量最大化和降低接收环境噪声。 由于海平面是强反射界面,在激发和接收环节都会产生虚反射效应,从而压制了信号的低频和高频能量,并产生了陷波点,限制了地震勘探的频带宽度。例如,为了获得深部目的层有效反射信号,必须增加气枪阵列容量、加大沉放深度以得到穿透能力大、主频低的激发子波,并加大电缆沉放深度以减少对来自深部反射界面的低频反射信号的压制效应,由此带来的副作用是高频信号受到较大压制,降低了地震信号的频带宽度和分辨率。 在海洋高分辨率地震勘探中,一般采用较小气枪阵列容量和较浅沉放深度以得到高频成分丰富的激发子波,同时降低电缆沉放深度以降低接收环节对高频信号的压制效应,这样虽然提高了地震信号的频带宽度和视觉分辨率,但它是以牺牲低频信息和勘探深度为代价,处理后的成果数据缺少低频信息,给后续的反演处理带来较大困难。 勘探设备性能也限制海洋地震勘探获得宽频带地震数据的能力,电缆在移动时产生的机械和声波噪声掩盖了微弱的有效地震信号,降低了地震数据的频宽和信噪比,尤其是对高频段信号的影响幅度更大。到目前为止,常规海洋地震勘探中尚未找到完全有效压制虚反射效应的采集和处理方法。 近年来,针对海洋宽频带地震勘探面临的主要难题,在勘探设备方面进行了研发并取得重要进展。固体电缆的研制成功和工业化应用,有效地降低了电缆噪声,提高了对微弱高频信号的响应和记录能力;双检波器拖缆采集技术的发展与应用,压制了虚反射效应,拓宽了地震频带。 众所周知,气枪和电缆以一定深度沉放于海平面之下,海平面反射在上行波和下行波之间产生交互干涉的鬼波效应,对地震反射信号产生了压制和陷波作用,降低了原始地震资料的频带宽度。气枪和电缆沉放越深,对高频信号压制越大,越有利于低频信号;沉放越浅,对低频信号压制越大,越有利于高频信号。 为了压制虚反射效应,提高地震数据频带宽度,在海洋地震激发时借鉴陆上地震勘探压制虚反射的成功做法,开发了多层震源组合新技术代替传统的平面震源组合方式,激发地震子波的低频和高频分量都得到有效拓展和提升,因此其频带展宽、穿透能力增强。 在海洋地震信号接收环节,为有效削弱由海平面虚反射引起的陷波作用,利用电缆沉放深度的变化对不同频带的压制特性,采用上、下缆接收技术,既有效

英语课题申报表版

编号: 淮安市教育科学“十二五”规划 课题申报评审书 课题名称:在新课标下,有关分层教学 的英语课堂模式的研究 研究方向与范围:教学方式与学习方式变革的研究 课题主持人:xxx翔 所在单位:xxx学校 申报日期:2014年12 月 1 日 ﹡淮安市教学研究室制﹡

填报说明 1.淮安市教育科学“十二五”规划课题申报者填写本《申报评审书》。 2.填写时,态度要认真负责,内容要实事求是,表达要严谨明了,字迹要清晰易辨。可以直接上市教研室网站下载本《申报评审书》后在电脑上填写,也可下载后用钢笔填写。 3.封面左上角“编号”栏,所有申报者均无须填写,评审时由市教研室填写。“研究方向与范围”栏按本省《“十二五”教育科学规划课题指南》中的分类填写,如确实有必要,“课题主持人”可以填两人。 4.“课题研究人员基本信息”栏中,“课题主持人”相关信息以第一主持人为准。“淮安市教育科学‘十一五’规划课题完成情况”分以下几种:A.主持并已结题;B.主持但未结题;C.参与研究;D.未参与研究。申报者只须在“淮安市教育科学‘十一五’规划课题完成情况”栏填上序号“A、B、C、D”即可。“其它教育科研课题完成情况”栏须如实填写主持或参与研究的一至二项课题名称、课题级别及完成情况,未参与任何课题研究者如实填写“未参与”。 5.《申报评审书》打印或复印时一律用A4纸,于左侧装订成册,按规定的申报程序报送。市《申报评审书》一式三份原件上报。 6.《申报评审书》中“课题研究设计与论证报告”总字数不宜超过5000字,各栏目空间填写时可根据实际需要调节。 淮安市教学研究室地址:淮安市健康西路31号,邮编:223001;电话:83641293,83652098。

申报课题指南序号

申报课题指南序号 中国法学会“深入研究党的 十八届四中全会精神”重点专项课题 申请书 课题名称 课题主持人 主持人所在单位(公章) 中国法学会 2014年11月印制

申请人的承诺: 本人郑重声明该申请系自愿提出,本人对所填写的各项内容的真实性负责,保证没有知识产权争议。 本人已经认真阅读并完全接受《中国法学会“深入研究党的十八届四中全会精神”重点专项课题申报公告》、《中国法学会部级法学研究课题管理办法》的规定。如获准立项,本人承诺接受本表、上述申报公告和课题管理办法为有法律约束力的协议,遵守中国法学会的规定,按时完成研究任务,取得预期成果。 本人已经了解项目最终成果需进行鉴定,通过鉴定后予以结项。中国法学会有权使用本项目的研究成果(包括阶段性成果和最终成果)。 课题申请人(亲笔签名): 年月日 课题申请人所在单位:(盖章) 年月日

填表说明 一、请严格按照本表格式填写,可加行,但勿随意变动字体字号、格式。 二、请用计算机认真如实填写(手写无效),如有不实则取消申报资格。 三、本表所有栏目均应如实填写,填写不完整、不按要求填写的申请表有可能被视为无效申请。 四、表中有选择项的请直接在该选项前的字母上划“√”。 (一)工作单位:应填写申请人档案关系所在单位全称,如中国人民大学法学院。 (二)地址:须填写详细通讯地址(不能仅以单位名称代替)和邮政编码。 (三)课题组成员名单:指除项目主持人外的其他项目参加人,不包括科研管理、财务管理、后勤服务等人员。 (四)凡用括号标注的,直接填写括号中的内容即可,如课题组成员一栏中,(姓名),直接填写课题组成员的名字,如李明。 (五)成果形式分为决策咨询报告(包括阶段性报告)、研究报告、论文、专著等。 五、申请人及课题组成员资格证明提供复印件即可,非A4纸大小的需粘贴在A4纸上,并装订在表后。 联系人:中国法学会研究部杨青春、曹菲 联系电话: (010)66173342 Email: clskt2014@https://www.wendangku.net/doc/417127319.html,

地震解释技术现状及发展趋势

第21卷 第2期地 球 物 理 学 进 展V ol.21 N o.22006年6月(页码:578~587) P ROG RESS IN G EOP HY SICS June. 2006 地震解释技术现状及发展趋势 张进铎 (东方地球物理公司研究院,涿州072751) 摘 要 本文以我国塔里木油田石油地球物理勘探实例为基础,概述了石油勘探过程中地震解释技术类型、特征、现状和发展趋势.本文认为,在地震勘探技术飞速发展的今天,地球物理学家及地质学家希望获得的地震信息,应当是能够直接反应地下岩石物理特性或油气水的分布,而利用常规的地震解释技术是很难做到这些;随着石油勘探的进一步深化,一些新的地震解释技术涌现出来,并在油气勘探与开发过程中发挥着巨大作用.未来的石油勘探将会面临前所未有的困难,新情况、新问题将层出不穷,地震解释技术也同样面临着考验,因此,只有立足在现有的成熟解释技术之上,并不断探索新的技术与思路,才能与未来的石油勘探步伐相一致. 关键词 塔里木盆地,地震相干技术,地震相分析技术,波阻抗反演技术,三维可视化技术中图分类号 P631 文献标识码 A 文章编号 1004-2903(2006)02-0578-10 Present status and future trend of seismic data interpretation techniques ZH ANG Jin -duo (Geoph ysical R esearc h Institute ,BGP ,CN PC,Zh uoz hou 072751,China) Abstract Dur ing the o il and gas explorat ion techniques develo ping ,g eophysicists and g eolo gists ho pe to use the seis -mic data to recog nize the ro ck features and o il and water dist ributions,it is difficult to do these by using the o rdinary seism ic data inter pr etatio n techniques.A few new techniques have been used with the oil and gas ex plor ation dev elop -ments,and t hese techniques have a lot of adv ant ages in practical a pplies.O il and g as ex plo ratio n w ill be faced w ith many difficulties in t he future,new co nditions and new pr oblems w ill be generated,the seismic dat a interpretation techniques w ill be also faced w ith new tests,so,w e must use mature techniques now ,and at the same time,dev elo ping new techniques and methods to match the steps in the future o il and gas ex plor ation. Keywords T ar im basin,seismic co her ent technique,seismic face technique,seismic inv ersion t echnique,3D v isualiza -t ion technique 收稿日期 2005-04-10; 修回日期 2005-08-20. 作者简介 张进铎(1966-),男,河北徐水人,硕士学位,高级工程师,从事三维地震解释与地震解释新技术应用工作.地址:河北省涿州市 贾秀路东方地球物理公司研究院总工办.(E -mail:peter_zhang@https://www.wendangku.net/doc/417127319.html,) 0 引 言 近年来,随着科学技术的迅速发展,在石油、天然气勘探领域中,地震资料解释和地质综合研究技 术有了飞速发展,新技术新方法层出不穷,以地震相干解释技术[1~5]、地震相分析技术[6~8]、波阻抗反演 技术[9,10]、三维可视化解释技术[11] 等为代表的一系 列新的地震解释技术[12] 在实际工作中得到了全面推广应用和发展. 现今的地震资料解释已不仅仅满足于常规的构造解释,它更倾向于以地震信息为主,借助先进的解 释技术,开展储层特征综合分析、油气藏分布规律等 更深层次的研究. 1 目前主要地震解释技术类型和现状 1.1 地震相干解释技术 地震相干解释技术[13]就是利用地震波形相干原理,计算中心地震道和指定相邻道的相干系数,将普通地震资料转换成相干系数资料,以突出地震资料中的异常现象. 该技术能快速建立起断裂系统、特殊岩性体的空间展布形态,指导岩性体和断层的剖面解释及平

最新研究专项课题指南

山西省“1331工程”研究专项课题指南 2017年11月

山西省“1331工程”研究专项课题指南 一、“1331工程”战略研究(14项) 双一流建设视域下中西部高等教育发展战略研究 双一流战略对地方院校办学定位的影响研究 地方高校双一流建设的政策机遇与路径选择 “1331工程”与山西高等教育发展路径选择研究 “1331工程”背景下高教资源优化配置与改革效益提升研究 “1331工程”与放管服改革、高校综合改革政策资源整合机制研究 “1331工程”战略实施多部门协同联动机制研究 “1331工程”对资源型省份创新驱动战略的支撑作用研究 “1331工程”的财政资金使用效益优化研究 “1331工程”战略中地(市)政府的响应与支撑作用研究 “1331工程”项目绩效评价机制研究 “1331工程”奖惩机制研究 “1331工程”政策效能评估研究 “1331工程”与山西农谷战略互动机制研究 二、立德树人工程研究(10项) 立德树人根本任务对高校办学战略引领作用研究 立德树人大协同机制构建研究 山西省重点马克思主义学院建设研究

山西省多类型协同育人机制构建研究 立德树人特色化品牌项目创建研究 立德树人协同育人队伍建设研究 立德树人工程综合效益评价研究 新时代立德树人思政课程与课程思政协同研究 新时代立德树人政工干部队伍胜任力研究 立德树人战略背景下辅导员团队化建设研究 三、重点学科建设研究(10项) 山西省重点学科建设的困境与对策研究 山西省重点学科建设制度创新研究 高校重点学科建设的模式、问题与对策 高校重点学科建设资源整合研究 高校重点学科建设投入产出效益评价研究 重点学科带头人成长规律与培养机制研究 重点学科赶超战略研究 学科集群服务产业集群战略研究 新升博硕学科建设后发战略研究 学科建设动态调整机制对高校学科优化的影响研究四、重点实验室建设研究(10项) 山西省高校重点实验室运行机制调查研究 山西省重点实验室运行模式、问题与对策研究 重点实验室管理理念与机制创新研究 重点实验室绩效综合评价研究 重点实验室建设障碍性因素与破解研究 重点实验室建设与复合型人才培养研究 重点实验室建设与创新型人才培养模式研究

地震勘探技术新进展_杨勤勇

第25卷第1期2002年2月 勘探地球物理进展 Progress in Exploration Geophysics Vol.25,No.1 Feb.2002地震勘探技术新进展 杨勤勇1徐丽萍2 (1.中国石化石油勘探开发研究院南京石油物探研究所,江苏南京210014; 2.西北石油局规划设计研究院,乌鲁木齐830011) 摘要:近几年来,地震勘探技术得到了很大的发展。超万道地震仪的投入使用,以及优化采集设计技术的发展,有效地提高了采集效率和资料质量;叠前深度偏移技术使复杂构造的成像更为清晰;3D可视化技术和虚拟现实技术大大提高了地震解释的能力、精度和速度;地震属性技术的发展把地震解释向定量化解释推进了一步;井中地震技术、多波多分量地震技术以及时延地震技术的发展,有力地增强了油气静态描述和动态监测的能力;复杂介质中地震波传播规律的研究向传统的层状介质理论发起了冲击。 关键词:可视化;虚拟现实;地震属性;成像;井中地震;VSP;多分量;时延地震 中图分类号:TE132.1+1文献标识码:A 地震勘探是利用地层岩石的弹性特性来研究地下地质结构,推断岩体物性,预测油气的一种勘查方法。几十年来,地震勘探以其高信噪比、高分辨率、高保真度、高精确度、高清晰度和高可信度等赢得了广大用户的信任,成为找油找气的关键技术。在油气勘探开发中,应用地震勘探已有效地解决了一系列复杂的地质问题,在各种复杂构造油气藏和隐蔽油气藏的勘查方面取得了重大成果,给油气公司带来了可观的经济效益。 近几年来,以PC计算机群大规模投入使用,可视化、虚拟现实、网络技术飞速发展为标志,以高分辨率地震、3D地震为代表,以4D地震、井中地震、多波多分量地震为发展前沿的地震勘探技术正跃上新的台阶,高密度采集和3D空间成像归位技术以其精确、灵活显示等优点,在国内外已卓有成效地用于查明各种复杂构造油气藏和隐蔽油气藏。 1主要进展 1.13D可视化技术[1~4] 可视化技术是把描述物理现象的数据转化为图形、图像,并运用颜色、透视、动画和观察视点的实时改变等视觉表现形式,使人们能够观察到不可见的对象,洞察事物内部结构。方法包括以图形为基础(或称为面可视化)和以体素为基础(体可视化)的可视化。在以体素为基础的体可视化中,每一个数据采样点被转换成一个体素(一个3D象素的大小近似于面元间隔和采样间隔)。每一个体素有一个对应于源3D数据体的值,一个RGB(红色、绿色、蓝色)色彩值以及可被用来标定数据透明度的暗度变量。 多年来,许多公司致力于地学可视化应用软件的开发,取得了可喜的成果。在3D图形工作站环境支持下,各种基于数据体操作、图素提取与曲面造型、体绘制技术的应用软件相继出现,它们基本上代表了当今综合解释工作站3D可视化软件功能的发展水平(见表1)。 表1有代表性的可视化解释处理软件 公司软件 Landmark 3DVI(3D体积解释) Voxcube(3D立体动画) Geoquest GeoViz(交互3D解释) Paradigm Voxel Geo(真3D地震解释系统) DGI Earth Vision (基于3D空间地质建模) Photo3DViz(3D体可视化) 体可视化允许解释人员直接进行地层解释,识别地震相,改进油藏特征描述。它通过数据的3D 立体显示,使解释人员能够作构造、断层、地层沉积、岩性、储集参数和油气等的交互解释。解释结果在三度空间内立体显示,可以激发资料处理解释人员的科学灵感,赋予他们无限的想像空间与创造力,极大地提高了工作效率和工作质量。 1.2虚拟现实技术 虚拟现实(Virtual Reality,简写为VR)是一种 收稿日期:2001-12-31 作者简介:杨勤勇(1964-),高级工程师,1985年毕业于中国地质大学物探系,现从事情报研究。

相关文档