1,995
views
1
recommends
+1 Recommend
2 collections
    3
    shares

      Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Cytotoxic compounds from marine actinomycetes: sources, structures and bioactivity

      review-article
      Bookmark

            Abstract

            Marine actinomycetes produce a substantial number of natural products with cytotoxic activity. Actinomycete strains have been isolated from sources including fishes, coral, sponges, seaweeds, mangroves and sediments. These cytotoxic compounds can be broadly categorized into four classes: polyketides; non-ribosomal peptides and hybrids; isoprenoids and hybrids; and others, among which the majority are polyketides (146 of 254). Twenty-two of the 254 compounds show potent cytotoxicity, with IC50 values at the ng/mL or nM level. This review highlights the sources, structures and antitumor activity of 254 natural products isolated from marine actinomycetes and first reported between 1989 and 2020.

            Main article text

            1. INTRODUCTION

            The oceans occupy more than two-thirds of the Earth’s surface and contain more than four-fifths of the world’s plant and animal species, in addition to a vast number of microorganisms [1]. Marine natural products usually refer to compounds isolated from marine microorganisms and phytoplankton, algae, sponges, cnidarians, bryozoans, mollusks, tunicates, echinoderms, mangroves, and other intertidal plants and microorganisms [2]. The discovery rate of new marine natural products has increased since the advent of this field and has continued at a substantial rate despite the ever-increasing number of reported natural products [3]. To date, 17 marine-derived drugs have been approved for clinical use: cytarabine (Cytosar-U®), vidarabine (Arasena A®), ziconotide (Prialt®), eicosapentaenoic acid ethyl ester (Vascepa®), omega-3-carboxylic acid (Epanova®), omega-3-acid ethyl esters (Lovaza®, whose status is currently debated), eribulin mesylate (E7389, Halaven®), trabectedin (ET-743, Yondelis®), panobinostat (Farydak®️), plitidepsin (Aplidin®), lurbinectedin (Zepzelca™), belantamab mafodotin-blmf (Blenrep™), brentuximab vedotin (SGN-35, Adcetris®), polatuzumab vedotin (DCDS-4501A, Polivy™), enfortumab vedotin-ejfv (PADCEV™), disitamab vedotin (Aidixi™) and tisotumab vedotin-tftv (Aidixi™) [4]. More marine natural products are highly likely to be developed for clinical use. Some of the lead compounds developed into the above-mentioned clinical drugs are likely to be produced by microorganisms including actinomycetes, given the growing recognition in recent decades that metabolic processes in microorganisms including actinomycetes are the most productive source of unique secondary metabolites [5]. Actinomycetes are a diverse family of filamentous bacteria that produce a plethora of natural products with relevance to agriculture, biotechnology and medicine, including most antibiotics approved by the U.S. Food and Drug Administration [6]. In the 1940s, actinomycetes were first discovered for their antibiotic functions [7]. Subsequently, secondary metabolites of actinomycetes were widely exploited as antitumor drugs in the pharmaceutical industry. Several anticancer drugs have been developed from enediynes, such as gemtuzumab ozogamicin (Mylotarg®) and inotuzumab ozogamicin (Besponsa®) [8, 9]. However, the high toxicity, undesirable adverse effects and extensive drug resistance of current treatments have increased the demand for novel antitumor drugs. Marine actinomycetes are a valuable source of biologically active secondary metabolites. According to a statistical analysis, marine-derived actinomycetes account for the production of 39% of all bioactive microbial metabolites [10]. This review describes the sources, chemical structures and cytotoxic activities of 254 compounds derived from marine actinomycetes, reported from 1989 to 2020.

            2. STRUCTURAL CLASSES OF ANTITUMOR SECONDARY METABOLITES FROM MARINE ACTINOMYCETES

            2.1 Polyketides
            2.1.1 Macrolides (lactones), lactams and α/γ-pyrones

            Two new kijanimicin derivatives, lobophorins C (1) and D (2), have been purified from Streptomyces carnosus AZS17 ( Figure 1 ). Compounds 1 and 2 each have a unique β-keto-γ-spiro-γ-lactone moiety with a double bond at the α-position. Compound 1 showed cytotoxicity toward the 7402 human liver cancer cell line and MDA-MB 435 human breast cancer cells with IC50 values of 0.6 μg/mL and 61.8 μM, respectively, while compound 2 was toxic against the same two cancer cell lines with IC50 values of 723.1 μg/mL and 7.5 μM, respectively [11]. Octalactin A (3), a fully saturated eight-membered lactone, has been isolated from a marine bacterium, Streptomyces sp. PG-19. Compound 3 displays inhibitory activity toward the B-16-F10 and HCT-116 cell lines, with IC50 values of 7.2 and 500 ng/mL, respectively [12]. The macrolides PM100117 (4) and PM100118 (5) have been obtained from Streptomyces caniferus GUA-06-05-006A [13]. Compounds 4 and 5 each have a 36-membered macrolide ring system together with three six-membered hemiketal rings and a naphthoquinone moiety on the bulky tail. Compound 4 displays cytotoxicity toward A549, MDA-MB-231 and HT29 cell lines, with GI50 values of 1.3, 2.7 and 3.8 μM, respectively, and compound 5 is active toward these three cell lines, with GI50 values of 0.83, 1.7 and 9.2 μM, respectively. A 16-membered diene macrolide, bafilomycin M (6), has been obtained from Streptomyces sp. GIC10-1, which has been isolated from the Theonella marine sponge species [14]. Compound 6 exhibits potent anticancer activity toward HL-60, SUPT-1, K-562 and LNCaP cells, with IC50 values of 11, 47, 60 and 389 ng/mL, respectively. The cytotoxic bafilomycin analogs bafilomycins N (7) and O (8) have been obtained from Streptomyces sp. GIC10-1 [15]. Compound 7 is cytotoxic to LNCaP, SUP-T1, MOLT-4 and K562 cells, with IC50 values of 3.9, 6.0, 0.01 and 31.8 nM, respectively, and compound 8 is active toward the same cancer cell lines, with IC50 values of 118.6, 64.4, 389.6 and 54.2 nM, respectively. The new compound lobophorin K (9) has been separated from the culture of Streptomyces sp. M-207 isolated from the cold-water coral Lophelia pertusa [16]. Compound 9 exhibits cytotoxicity toward THLE-2, MCF-7 and MiaPaca-2, with IC50 values of 6.3 ± 8.2, 23.0 ± 8.9 and 34.0 ± 85.1 μM, respectively. A new spirotetronate lobophorin F (10) has been isolated from Streptomyces sp. SCSIO 01127 and shown activity toward the SF-268, MCF-7 and NCI-H460 cell-lines, with IC50 values of 6.82, 2.93 and 3.16 μM, respectively [17].

            Figure 1 |

            Structures of compounds 1–27.

            The new α-pyrone derivatives violapyrones H (11) and I (12) have been obtained from Streptomyces sp. isolated from the crown-of-thorns starfish, Acanthaster planci [18]. Compounds 11 and 12 are cytotoxic toward HCT-15, HCT-116, MDA-MB-231, NCI-H23, NCI-H460, NUGC-3, Hep-G2 and PC-3 cells, with IC50 values in the range of 1.10 to 25.05 μM. Nocardiopsis sp. NHF48 has been found to produce a new α-pyrone compound (13) exhibiting cytotoxic activity toward the melanoma cell line B16, with a GI50 value of 61.7 μg/mL [19]. From Streptomyces sp. HKI0576, ansa-macrolides divergolides A–D (14–17) have been obtained [20]. Compound (17) displays cytotoxicity toward lung cancer (LXFA 629L), pancreatic cancer (PANC-1), renal cancer (RXF 486L) and sarcoma (Saos-2) cells, with IC50 values in the range of 1.0 to 2.0 μM. Aureoverticillactam (18), a 22-atom macrocyclic lactam incorporating both triene and tetraene conjugated olefins, has been obtained from Streptomyces aureoverticillatus NPS001583, and has shown cytotoxicity toward HT-29, B16-F10 and Jurkat cells, with EC50 values of 3.6, 2.2 and 2.3 μM, respectively [21]. Two new 16-membered macrolides, 21,22-en-bafilomycin D (19) and 21,22-en-9-hydroxybafilomycin D (20), have been purified from the seaweed-derived Streptomyces sp. HZP-2216E [22]. Compound 19 displays cytotoxicity toward U251 and C6 glioma cell lines, with IC50 values of 1.08 and 0.21 μM, respectively, and compound 20 is toxic toward the same cell lines, with IC50 values of 0.36 and 0.12 μM, respectively. A 42-membered macrolide, desertomycin G (21), has been obtained from cultures of the marine actinomycete Streptomyces althioticus MSM3 isolated from samples of Ulva sp. intertidal seaweed collected in the Cantabrian Sea (Northeast Atlantic Ocean) [23]. Bioevaluation results have indicated that, at day 3, DLD-1 and MCF-7 cancer cell lines show a decrease in viability to approximately 50% that of controls after treatment with 2.5 and 5.0 μM desertomycin G (21). From a mangrove actinomycete strain, Streptomyces sp. 219807, which produces a high yield (4,486 mg/L) of elaiophylins, has been isolated [24]. A new elaiophylin derivative, halichoblelide D (22), has been obtained and identified from 219807 [24]. Compound 22 exhibits cytotoxic activity toward MCF-7 and HeLa cells, with IC50 values of 0.33 and 0.30 μM, respectively.

            Compound 23 is composed of four partial structures: cyclopenta[a]indene, 3′-chloro-5′-hydroxy-β-tyrosine, benzoxazine and amino sugar. Compound 23 shows cytotoxicity toward MDA-MB231, HCT-116, A549, SNU638, K562 and SK-HEP1, with IC50 values of 0.9, 2.7, 14.7, 9.8, 25.1 and 7.9 μM, respectively [25]. Compound 23 shows cytotoxicity toward MCF-7, with an IC50 of 27.0 μg/mL. Four new nonacyclic dilactones, antimycins E−H (24–27), have been obtained from Streptomyces sp. THS-55 [26]. Compounds 24–27 each contain a N-[3-(acetylamino)-2-hydroxybenzoyl] moiety. Compounds 24–26 are cytotoxic to Siha, K562, HL-60 and 293T cancer cell lines, with IC50 values of 0.8–13.8 μM. Two new benzamido nonacyclic dilactones, neoantimycins A (28) and B (29), have been obtained from Streptomyces antibioticus H12-15 ( Figure 2 ) [27]. The actinomycete strain S. antibioticus H12-15 has been isolated from a sea sediment in a mangrove district. Compounds 28 and 29 exhibit anticancer activity toward SF-268 cells, with IC50 values of 68.7 and 87.6 μM, respectively. Three new 4H-chromen-4-one polyketides, phaeochromycins F–H (30–32), have been separated from Streptomyces sp. DSS-18, a strain isolated from a deep-sea sediment collected from the western Pacific [28]. Compounds 30–32 are active toward HeLa cells, with inhibition rates of 9.4%, 1.0% and 46.0% at 10 μg/mL, respectively.

            Figure 2 |

            Structures of compounds 2865.

            Tartrolon D (33), a cytotoxic macrodiolide with two hemiketal rings, has been separated from Streptomyces sp. MDG-04-17-06 isolated by spreading a marine sediment collected near the east coast of Madagascar on 172B modified agar medium plates supplemented with nalidixic acid (1%) [29]. Compound 33 has cytotoxic activity toward A549, HT29 and MDA-MB-231 cells, with GI50 values of 0.16, 0.31 and 0.79 μM, respectively. Two new macrocyclic lactones, azalomycin F4a 2-ethylpentyl ester (34) and azalomycin F5a 2-ethylpentyl ester (35), have been separated from a culture of Streptomyces sp. 211726 isolated from a mangrove rhizosphere soil sample [30]. Compounds 34 and 35 are cytotoxic toward HCT-116 cells, with IC50 values of 5.64 μg/mL and 2.58 μg/mL, respectively. Seven new azalomycin F analogs (36–42) have been obtained from Streptomyces sp. 211726 [31]. These macrolides (36–42) display inhibitory activity toward HCT-116 cells, with IC50 values ranging from 1.81 to 5.00 μg/mL.

            Six new polycyclic tetramate macrolactams, pactamides A–F (43–48), have been purified from the marine-derived strain Streptomyces pactum SCSIO02999, and have shown cytotoxicity toward four cancer cell lines (MCF-7, SF-268, Hep-G2 and NCI-H460), with IC50 values ranging from 0.24 to 25.47 μM [32].

            Two new macrolides, pulvomycin B (49) and pulvomycin D (50), have been discovered from an estuarine Streptomyces strain [33]. Compound 49 displays cytotoxic activity toward HCT116, SNU638, SK-Hep-1 and MDA-MB-231 cells, with IC50 values in the range of 3.7−25 μM, whereas compound 50, which bears a 1,2-diketone functional group, strongly inhibits the same cancer cell lines, with IC50 values ranging from 0.21 to 0.40 μM. A new curvularin glycoside, curvularin-7-O-α-D-glucopyranoside (51), has been isolated from Pseudonocardia sp. HS7 obtained from the cloacal aperture of the sea cucumber Holothuria moebii [34]. Compound 51 displays inhibitory activity against all six cancer cell lines (HCT-15, C6, U251, SHG-44, U87-MG and SW620), with IC50 values in the range of 20.84 to 81.01 μM. Three new polyene macrolactams, kenalactams C−E (52–54), have been separated from Nocardiopsis CG3 (DSM 106572) isolated from the saltpan of Kenadsa [35]. Compounds 52 and 53 show cytotoxicity toward L929, KB3.1, MCF-7, PC-3, A549 and SKOV-3 cells, with IC50 values ranging from 5.4 to 42.2 μM. Compound 54 is also active toward KB3.1, PC-3, SKOV-3 and A549 cells, with IC50 values ranging from 2.1 to 6.5 μM. Cultivation of Micromonospora sp. FIM05328 has yielded the macrolactam FW05328-1 (55) [36].

            Compound 55 inhibits the KYSE30 and KYSE180 tumor cell lines, with IC50 values of 15.92 and 30.77 μM, respectively. Interestingly, compound 55 strongly inhibits the esophageal cancer EC109 cell line, with an IC50 value of 0.2 nM. The Micromonospora strain FIM07-0019 has yielded a new 20-membered macrolide, levantilide C (56) [37]. The strain FIM07-0019 has been recovered from shallow coastal water near the island of Chiloe, Chile. Compound 56 displays inhibitory activity toward HL-60, MDA-MB-231, SW620 and SMMC7721 cells, with IC50 values of 32.5, 26.8, 16.4 and 39.9 μM, respectively.

            A polycyclic tetramate macrolactam, 16-hydroxymaltophilin (57), isolated from Actinoalloteichus cyanogriseus WH1-2216-6, shows cytotoxicity toward BXPC-3, HCT-116, Jurkat, PANC-1, A549, MCF-7 and L-02 cell lines, with IC50 values of 4.5, 5.7, 7.5, 7.9, 9.5, 9.7 and 235.9 μM, respectively [38].

            2.1.2 Benzoquinones, naphthoquinones, anthraquinones and other aromatic compounds

            One anthracycline, tetracenoquinocin (58), has been separated from a culture of Streptomyces sp. Sp080513GE-26 associated with a Haliclona sp. marine sponge [39]. Compound 58 is cytotoxic toward HeLa and HL-60 cells, with IC50 values of 120 and 210 μM, respectively. The new salicylamide derivative JBIR-58 (59) has been obtained from Streptomyces sp. SpD081030ME-02 isolated from a demospongiae class of marine sponge [40]. Compound 59 displays inhibitory activity toward HeLa cells, with an IC50 value of 28 μM. Streptomyces sp. HB202 has been found to produce the new benz[a]anthracene derivative mayamycin (60), which displays cytotoxicity toward HepG2, MAXF401NL, MEXF462NL, HT-29, GXF251L, LXF529L, PAXF1657L and RXF486L cells, with IC50 values ranging from 0.13 to 0.33 μM [41]. Streptomyces sp. BCC45596 has yielded three new C-glycosylated benz[a]anthraquinone derivatives: urdamycinone E (61), urdamycinone G (62) and dehydroxyaquayamycin (63) [42]. Compounds 61 and 62 display inhibitory activity toward KB, MCF-7 and NCl-H187 cancer cell lines, with IC50 values ranging from 0.092 to 0.45 μg/mL, whereas compounds 63 is much less active toward these three cancer cell lines, with IC50 values of 6.96, 3.41 and 3.97 μg/mL, respectively. All three compounds (61–63) are less toxic toward non-cancerous (Vero) cells than cancer cells, with IC50 values of 1.71, 3.05 and 10.07 μg/mL, respectively. Three angucyclinone derivatives, monacyclinone C (64), monacyclinone E (65) and monacyclinone F (66; Figure 3 ), have been purified from Streptomyces sp. M7_15 associated with the sponge Scopalina ruetzleri, which displays inhibitory activity toward SJCRH30 cells, with IC50 values of 160, 270 and 0.73 μM, respectively [43]. The potent anticancer activity of compound 66 might be due to the two unique epoxide rings attached to the angucyclinone moiety. The chlorinated strepchloritides A (67) and B (68) have been separated from the oligotrophic culture of a soft coral-associated actinomycete strain, Streptomyces sp. OUCMDZ-1703, and have shown cytotoxicity toward MCF-7 cells, with IC50 values of 9.9 and 20.2 μM, respectively [44]. Naquihexcin A (69), an S-bridged pyranonaphthoquinone dimer bearing an unsaturated hexuronic acid moiety, has been obtained from the sponge-derived Streptomyces sp. HDN-10-293 [45]. Compound 69 is cytotoxic toward MCF-7 ADM, with an IC50 value of 16.1 μM. A coral-derived strain, Streptomyces sp. RKBHB7, produces a new meroterpenoid with a sesterterpene skeleton, guanahanolide A (70), with cytotoxicity toward MCF-7, NCI-60, HCT-116, HTB-26 and Vero cells, with IC50 values of 7.8, 10.0, 11.9, 10.1, 23.7 μM, respectively [46]. Streptomyces sp. ZZ406 has yielded l-hydroxymethyl-8-hydroxy-anthraquinone-3-carboxylic acid (71) and a 2-methylchromone derivative, phaeochromycin I (72) [47]. Compound 71 displays inhibitory activity toward the glioma cells U251, U87MG and SHG, with IC50 values of 5.7, 4.7 and 8.1 μM, respectively, whereas compound 72 is less active toward U251, U87MG and SHG glioma cells, with IC50 values of 21.6, 25.7 and 25.8 μM, respectively. Streptomyces sp. CANU Fox 21-2-6a, isolated from the outer layer of driftwood material collected at the mouth of the Fox River on the West Coast of New Zealand, has yielded four new anthracycline derivatives: (7S*9R*10R*)-pyrromycin (73), (7R*9R*10R*)-pyrromycin (74), 1-hydroxyauramycin T (75), and 1-hydroxysulfurmycin T (76) [48]. Compounds 73–76 are cytotoxic toward the P388 tumor cell line, with ID50 values in the range of 0.4–0.06 μg/mL. A new anthraquinone, 1,8-dihydroxy-2-ethyl-3-methylanthraquinone (77), has been separated from a fermentation of Streptomyces sp. FX58-1 isolated from the marine plant Salicornia herbacea collected in Qingdao, Shandong province, China [49]. Compound 77 is cytotoxic toward HL-60, BCTC-823 and MDA-MB-435 cells, with IC50 values of 6.83, 82.2 and 56.59 μg/mL, respectively. A culture of Streptomyces sp. B8652 has been found to produce parimycin (78) [50]. The strain B8652 has been isolated from a sediment of the Laguna de Terminos at the Gulf of Mexico. Compound 78 is cytotoxic toward LXFA629L, LXFL529L, MCF-7, MAXF401NL, MEXF462NL and MEXF 514L cells, with IC50 values in the range of 0.9–6.7 μg/mL. Fermentation of Streptomyces sp. M045 derived from a sediment collected at Jiaozhou Bay in China has led to the identification of chinikomycins A (79) and B (80) [51]. Compound 79 is a hydroquinone derivative, whereas compound 80 is a 1,4-benzoquinone analog, which might be oxidized from 79. Compound 79 is cytotoxic toward MAXF 401NL, MEXF 462NL and RXF 944L cells, with IC50 values of 2.41, 4.15 and 4.02 μg/mL, respectively, and compound 80 is active toward MAXF 401NL cells, with an IC50 value of 3.04 μg/mL. Two anthraquinones of the angucycline class, marmycins A (81) and B (82), have been obtained from Streptomyces sp. CNH990 [52]. Compound 81 is a monochloro derivative of compound 82. Compounds 81 and 82 show cytotoxicity toward HCT-116 cells, with IC50 values of 60.5 nM and 1.09 μM, respectively. Compound 81 shows cytotoxicity toward 12 human tumor cell lines (lung, colon, breast, prostate or leukemia) after 72 h drug exposure, with IC50 values ranging from 7 to 58 nM, but compound 82 shows cytotoxicity toward the above 12 human tumor cell lines, with IC50 values ranging from 1.0 to 4.4 μM. The results contrast with general observations that chlorination usually markedly enhances the pharmacological activity of compounds [53]. Three new anthramycin-type analogues, usabamycins A–C (83–85), have been purified from Streptomyces sp. NPS853, a bacterial strain found in a marine sediment [53]. Compounds 83–85 are pyrrolo[1, 4]benzodiazepine derivatives that display weak inhibitory activity toward HeLa cells, with IC50 values of 106.6, 103.5 and 101.9 μM, respectively. A new anthracene derivative, 3-hydroxy-1-oxo-3-methyl-8-methoxy-1,2,3,4-tetrahydro-benz[α]anthracene (86), has been isolated from the fermentation broth of Streptomyces sp. W007 [54]. In cytotoxicity tests, compound 86 shows no cytotoxicity toward the human leukemic cell line HL-60 and weaker cytotoxicity toward the human hepatoma cell line BEL-7402 than adriamycin, but potent inhibitory activity toward the human lung adenocarcinoma cell line A549, with a rate of inhibition at 1 μM of 61.8%. Four angucycline C-glycosides, grincamycins B−E (87–90), have been isolated from Streptomyces lusitanus SCSIO LR32, an actinomycete of deep-sea origin [55]. The disaccharide moiety in compound 89 forms a 1,4-dioxane ring through 3-2’ and 4-1’ linkages. Compounds 87–90 show cytotoxicity toward MCF-7, HeLa, HepG2, B16, NCI-H460 and SW-1990 cells, with IC50 values in the range of 2.1 to 31 μM. Streptomyces sp. SNE-011 has yielded the arylamine derivatives carpatamides A (91) and C (92) [56]. Strain SNE-011 has been isolated from a marine sediment sample collected from South Carolina. Compound 91 exhibits inhibitory activity toward HCC366, A549 and HCC44 cells, with IC50 values of 2.8, 4.1 and 8.4 μM, respectively, and compound 92 inhibits HCC366 and A549 cells, with IC50 values of 2.2 and 3.7 μM, respectively. The compound (2S,3R)-L-threonine, N-[3-(formylamino)-2-hydroxybenzoyl]-ethyl ester (streptomyceamide C, 93) has been isolated from EtOH extract of the fermented mycelium of the marine-derived streptomycete strain H74-21 isolated from a sea sediment in a mangrove site [57]. Compound 93 shows cytotoxicity toward MCF-7, with an IC50 of 27.0 μg/mL. Deep-sea sediment-derived Streptomyces sp. SCSIO 11594 has yielded an angucycline C-glycoside, marangucycline B (94), which displays cytotoxic activity toward A549, CNE2, MCF-7, HepG2 and HL7702 cells, with IC50 values of 0.45, 0.56, 0.24, 0.43 and 3.67 μM, respectively [58]. Compound 94, with a keto-sugar moiety and a 1,4-dioxane ring between sugars, is approximately 10–20-fold more potent than cisplatin. Elmonin (95) and elmenol B (96) have been separated from Streptomyces sp. IFM11490 and shown cytotoxicity toward the human gastric adenocarcinoma (AGS) cell line, with almost equal IC50 values of approximately 50.0 μM [59]. Cultivation of Streptomyces sp. 182SMLY has afforded two polycyclic anthraquinones, N-acetyl-N-demethylmayamycin (97) and streptoanthraquinone A (98) [60]. Compounds 97 and 98 display inhibitory activity toward C6, U251, U87-MG and SHG-44c cells, with IC50 values of 0.5/7.3, 0.7/3.3, 1.4/4.6 and 3.9/6.5 μM, respectively. The cell viability of normal human astrocytes from each tested concentration of both compounds 97 and 98 is 100%, and both compounds have IC50 values of 25 and 100 μM, respectively. Diazaquinomycin E (99) has been obtained from Streptomyces sp. F001 and has been found to display cytotoxicity toward OVCAR5, with an IC50 value of 9.0 μM [61]. A study of the Streptomyces griseus strain M268 has led to the identification of a unique cage-like compound, grisemycin (100), which is cytotoxic toward HL-60, with an IC50 value of 31.54 μM [62]. A novel dimeric diazobenzofluorene glycoside, lomaiviticin A (101), has been obtained from a halophilic strain, Micromonospora lomaiwitiensis LL-37I366 [63]. Compound 101 is a dimeric benzofluorene glycoside attached to two diazo functional groups at C-5 and -5′, which shows potent cytotoxic activity toward several cancer cell lines, with IC50 values in the range of 0.01 to 98 ng/mL. The compounds (9R,14S)-epoxy-11-deoxyfunicone (102) and (9S,14R)-epoxy-11-deoxyfunicone (103) have been obtained from co-cultivation of Streptomyces fradiae 007 and Penicillium sp. WC-29-5 [64]. A racemic mixture of enantiomers 102 and 103 has been separated with chiral chromatography. Compound 102 inhibits H1975 cells, with an IC50 value of 3.97 μM, and compound 103 inhibits HL-60 and H1975 cells, with IC50 values of 3.73 and 5.73 μM, respectively. Deep-sea-derived Streptomyces lusitanus SCSIO LR32 has been found to produce a new angucycline glycoside, designated grincamycin H (104), which is cytotoxic toward Jurkat T cells, with an IC50 value of 3.0 μM [65]. Two new angucycline glycosides, grincamycin I (105) and grincamycin J (106), are produced by marine-derived Streptomyces lusitanus SCSIO LR32 [66]. Compound 105 displays inhibitory activity toward MDA-MB-435, MDA-MB-231, NCI-H460, HCT-116, HepG2, and MCF10A cells, with IC50 values of 10.20, 25.87, 11.87, 8.79, 9.41 and 2.90 μM, respectively, and compound 106 is active toward the same cancer cell lines, with IC50 values of 2.63, 4.68, 5.40, 2.63, 4.80 and 2.43 μM, respectively. A culture of Streptomyces sp. XMA39 has afforded two medermycin-type naphthoquinones, strepoxepinmycins C (107) and D (108; Figure 4 ), which show cytotoxic activity toward HCT116 cells, with IC50 values of 4.4 ± 0.1 and 2.9 ± 0.1 μM, respectively [67]. Lagumycin B (109) has been discovered from Micromonospora sp. G039 [68]. Strain G039 has been isolated from a sediment sample collected by PONAR at a depth of 22 m, approximately 3.3 miles off the coast southeast of Cát Bà Peninsula in Vietnam. Compound 109 is cytotoxic to non-cancerous murine ovarian surface epithelial and murine oviductal epithelial cell lines, with LC50 values of 9.80 μM and 10.8 μM, respectively. Investigation of a bacterial strain from the South China Sea, Micromonospora echinospora SCSIO 04089, has led to the discovery of homophenanthroviridone (110), homophenanthridonamide (111) and nenesophanol (112) [69]. Compound 110 shows cytotoxicity toward the SF-268, MCF-7 and HepG2 cell lines, with IC50 values of 5.4 ± 0.4, 6.8 ± 0.3 and 1.4 ± 0.1 μM, respectively. Compound 111 is active toward these three cell lines, with IC50 values of 18 ± 1, 52 ± 2 and 4.0 ± 0.3 μM, respectively. Compound 112 is also active toward SF-268, MCF-7 and HepG2 cell lines, with IC50 values of 7.6 ± 0.9, 10.4 ± 0.5 and 8.1 ± 0.4 μM, respectively. Saccharothrix sp. 10-10 has yielded a new tetracenomycin analogue, saccharothrixone D (113), which exhibits cytotoxicity toward HepG2 cells, with an IC50 value of 7.5 μM [70]. Akazamicin (114), a new aromatic polyketide, has been obtained from the liquid culture of Nonomuraea sp. AKA32 was isolated from deep-sea water collected from a depth of 800 m in Sagami Bay, Japan, and compound 114 shows cytotoxicity toward murine B16 melanoma, Hep G2 and Caco-2 cells, with IC50 values of 1.7, 75 and 185 μM, respectively [71].

            Figure 3 |

            Structures of compounds 66–107.

            Figure 4 |

            Structures of compounds 108–148.

            2.1.3 Decalin derivatives

            Nahuoic acid A (115) has been obtained from Streptomyces sp. RJA2928 and found to inhibit SETD8 activity, with an IC50 value of 6.5 μM [72]. Nahuoic acids B–E (116–119) have been purified from the same strain, and 116–119 have been found to inhibit SETD8 activity with IC50 values of 27, 41, 76 and 13 μM, respectively [73]. The compound (1α,4aα,5α,7β,8aβ)-5,8a-dimethyl-decahydrona-phthalene-1,4a,7-triol (120) has been obtained from Streptomyces sp. 0616208 and has shown moderate inhibitory effects toward SMMC-7721 cells [74].

            2.1.4 Polyenes

            Piericidins C7 (121) and C8 (122) have been obtained from the culture of Streptomyces sp. YM14-060 isolated from unidentified greenish ascidians collected at Iwayama Bay, Palau [75]. Compounds 121 and 122 show cytotoxicity toward RG-E1A-7 and Neuro-2a cells, with IC50 values of 1.5 and 0.83, and 0.21 and 0.45 nM, respectively. A new nitro-tetraene spiro-β-lactone-γ-lactam, lajollamycin (123), has been isolated from Streptomyces nodosus (NPS007994) [76]. The strain NPS007994 has been isolated from a marine sediment sample collected at Scripps Canyons, La Jolla, California. Compound 123 exhibits cytotoxic activity toward the B16-F10 cell line, with an EC50 value of 9.6 μM. Piericidin F (124) has been isolated from the fermentation broth of Streptomyces sp. CHQ-64 and has shown cytotoxicity toward HeLa, NB4, H1975 and A549 cell lines, with IC50 values of 3, 37, 490 and 560 nM, respectively [77]. Four new pyranones, PM050511 (125), PM050463 (126), PM060054 (127) and PM060431 (128), have been isolated from the culture of Streptomyces albus POR-04-15-053. Compounds 125 and 128 show strong inhibition against MDA-MB-231, HT-29 and A549 cells, with IC50 values ranging from 0.24–0.69 μM [78]. Glucopiericidin C (129) has been isolated from an extract of a cultured Streptomyces sp. B8112 and found to display a concentration-dependent cytotoxicity toward a panel of 36 human tumor cell lines, with a mean IC50 of 2.0 μM (mean IC70 μM). [79]. Pterocidin (130) has been isolated and identified from Streptomyces sp. TP-A0879 isolated from a sediment sample collected at a depth of 44.5 m in Otsuchi Bay, Iwate, Japan by using the Smith–McIntyre grab method [80]. Compound 130 exhibits cytotoxic activity toward 26-L5 cells, with an IC50 value of 0.25 μM. Succinilene A (131) has been identified from Streptomyces strain SAK1 collected in the southern area of Jeju Island, Republic of Korea [81]. Compound 131 shows cytotoxicity toward SNU638 cells, with an IC50 value of 12.1 μg/mL (27.6 μM). The compound (2E,4Z,6E,8Z)-5,9-dimethyl-10-oxododeca-2,4,6,8-tetraenoic acid (132), a polyunsaturated acid, has been obtained from the liquid culture of Streptomyces violans HTTA-F0412 and has shown cytotoxicity toward A2780 cells, with an IC50 value of 4.36 μM [82]. Separacene A (133) has been isolated from Streptomyces sp. SNJ210 and found to display weak inhibitory activity toward HCT116 and A549 cells, with IC50 values of 14.0 μg/mL and 37.6 μg/mL, respectively [83].

            2.1.5 Other polyketides

            Daryamides A (134), B (135) and C (136), and (2E,4E)-7-methylocta-2,4-dienoic acid amide (137) have been discovered from Streptomyces sp. CNQ-085; these compounds exhibit cytotoxicity toward HCT116 cells, with IC50 values of 3.15, 9.99, 10.03 and 21.69 μg/mL, respectively [84]. Streptomyces sp. NPS-643 has yielded the tricyclic polypropionates indoxamycin A (138) and indoxamycin F (139), which exhibit cytotoxicity toward human colon adenocarcinoma HT-29 cells, with IC50 values of 0.59 and 0.31 μM, respectively [85]. Cyclizidines C (140) and D (141), each with a cyclopropane ring, have been isolated from Streptomyces sp. HNA39. Compound 140 shows cytotoxicity toward PC3, HCT116 and ROCK2 cells, with IC50 values of 0.52 ± 0.03, 8.3 ± 0.1 and 7.0 ± 0.8 μM, respectively. Compound 141 is much less active toward PC3 and HCT116 cells, with IC50 values of 33 ± 1 and 40 ± 1 μM, respectively [86].

            A new hydroxamate derivative, MBJ-0003 (142), has been isolated from Micromonospora sp. 29867 and has shown cytotoxic activity toward the SKOV-3 cell line, with an IC50 value of 11 μM [87]. Paulomycin G (143) has been discovered from Micromonospora matsumotoense M-412 isolated from Cantabrian Sea sediments collected at 2,000 m depth; this compound exhibits cytotoxicity toward pancreatic adenocarcinoma (MiaPaca_2), MCF-7 and HepG2 cells, with IC50 values of 2.70, 1.58 and 4.30 μM, respectively [88]. An extract of Verrucosispora sp. SCSIO 07399 has yielded three new kendomycin analogues, kendomycins B−D (144–146) [89]. Compounds 144–146 are macrocyclic polyketides, each containing a benzofuran-6(2H)-one connected to a tetrahydropyran moiety at C-4 of benzofuran-6(2H)-one. Compounds 144–146 show cytotoxicity toward MGC803, A549, HeLa, HepG2, MCF-7 and RKO cells, with IC50 values ranging from 2.2 to 44 μM.

            Among these 146 polyketides (1–146), compounds 3 [12], 6–8 [14], 55 [38], 73–76 [49], 81 [53], 101 [63], 121 and 122 [75], and 124 [77] show substantial cytotoxicity, with IC50 values at the ng/mL (or nM) level. Compounds 1, 2, 9, 10, 93, 101, 123, 140, 141 and 144–146 are structurally interesting. Notably, compound 101 is not only structurally unique but also cytotoxically potent. The structure of compound 101 is complex, and this molecule shows promise in anticancer drug development.

            2.2 Non-ribosomal peptides and hybrids of polyketides and peptides

            Streptomyces sp. LS298, obtained from the marine sponge Gelliodes carnosa, has produced a new analogue of echinomycin, quinomycin G (147), an octapeptide (Val-Cys-Ala-Ser-Val-Cys-Ala-Ser) cyclized between cysteine moieties with two quinoxalines attached to serine moieties [90]. Compound 147 shows cytotoxicity toward ACHN, 786-O, U87 MG, Jurkat, SW1990, Mia-PaCa-2, SK-N-SH, HCT-116 and NCI-H1650 cells, with IC50 values of 0.552, 0.721, 0.627, 0.414, 2.56, 4.75, 5.17, 8.16 and 3.90 μM, respectively. Streptomyces sp. SBT348 from the Mediterranean sponge Petrosia ficiformis has yielded a new cyclic dipeptide (hypogallate-Orn-Leu), petrocidin A (148), which exhibits cytotoxicity toward HT-29 and HL-60 cells, with IC50 values of 5.3 and 3.9 μg/mL, respectively [91]. Streptomyces sp. SNJ013 isolated from a deep-sea sediment collected off Jeju Island, Korea, has produced a new lasso peptide, sungsanpin (149; Figure 5 ) [92]. Compound 149 contains 15 amino acid units, composed of an eight-amino-acid macrocyclic ring (8-1Gly-Phe-Gly-Ser-Lys-Pro-Ile-Asp8-9) and a seven-amino-acid chain (8-9Ser-Phe-Gly-Leu-Ser-Trp-Leu15). Compound 149 displays inhibitory activity in cell invasion assays toward the human lung cancer cell line A549. The cyclic peptides ohmyungsamycins A (150) and B (151) have been found to be produced by Streptomyces sp. SNJ042 isolated from Shinyang Beach on Jeju Island [93]. Compounds 150 and 151 each contain 12 amino acid units with 10 amino acids in the ring (10-1Val-Phe-Val-Trp-Val-Val-Leu-Val-Thr-Thr10-11) and two on the side chain (11Val-Val12). Compound 150 exhibits cytotoxic effects against HCT116, A549, SNU-638, MDAMB-231 and SKHEP-1 cells, with IC50 values of 0.359, 0.551, 0.532, 0.688 and 0.816 μM, respectively. Compound 151 exhibits much less of a cytotoxic effect against the above-mentioned cells, with IC50 values ranging from12.4 to 16.8 μM. Both compounds 150 and 151 show virtually no cytotoxicity toward normal MRC-5 lung cells (IC50 > 40 μM), thus indicating that these compounds exhibit more selective antiproliferative activity toward cancer cells than normal cells. Streptomyces sp. P11-23B has afforded two cyclodepsipeptides, streptodepsipeptides P11A (152, cyclo-(D-Val-L-Lac-L-Val-D-Hiv- D-Val-L-Lac-L-Val-D-Hiv-D-Val-L- Lac-L-Val-D-Hba)) and P11B (153, cyclo-(D-Val-L-Lac-L-Val-D-Hiv-D-Val-L-Lac-L-Val-D-Hiv-D-Val-L-Lac-L-Val-L-Lac)) [94]. Compounds 152 and 153 are cytotoxic toward SHG-44, U87-MG, U251 and C6 cells, with IC50 values in the ranges of 0.3–0.4 and 0.1–1.4 μM, respectively. A new cyclic lipopeptide, iturin A6 (154), has been separated from Streptomyces sp. SSA 13 isolated from the Arabian Sea [95]. Compound 154 is a cyclic lipopeptide containing a C16 β-amino fatty acid chain attached to a hydrophilic heptapeptide ((fatty acid)CO-Asn-Tyr-Asn-Gln-Pro-Asn-Ser-NH(fatty acid)) of seven α-amino acids. Compound 154 displays cytotoxicity toward HeLa, MCF-7 and Hep-G2 cell lines, with IC50 values of 1.73 ± 0.9, 6.44 ± 0.6 and 8.9 ± 1.09 μg/mL, respectively. Three new 2,5-diketopiperazines, 3-(3-hydroxy-4-methoxybenzyl)-6-isobutyl-2,5-diketopiperazine (155), 3-(1,3-benzodioxol-5-ylmethyl)-6-isobutyl-2,5-diketopiperazine (156) and 3-(1,3-benzodioxol-5-ylmethyl)-6-isopropyl-2,5-diketopiperazine (157), have been obtained from Streptomyces sp. MNU FJ-36 [96]. All three new compounds, 155–157, exhibit cytotoxic activity toward A-549 cells, with IC50 values of 89.4, 35.4 and 28.4 μg/mL, respectively. Compounds 156 and 157 inhibit the growth of HCT116 cells, with IC50 values of 75.4 and 45.4 μg/mL, respectively. The compounds (S)-6-(sec-butyl)-3-Isopropylpyrazin-2(1H)-one (158) and (S)-6-(sec-butyl)-3-isobutylpyrazin-2(1H)-one (159) have been discovered from a tunicate-derived strain, Streptomyces sp. Did-27, and found to exhibit cytotoxicity toward HCT-166 cells with the same IC50 value of 30 μg/mL [97]. Compounds 158 and 159 show inhibitory effects toward MCF-7 cells, with IC50 values of 25 and 35 μg/mL, respectively. A new cyclic hexapeptide, nocardiotide A (160), has been isolated from the culture broth of Nocardiopsis sp. UR67 associated with the marine sponge Callyspongia sp. from the Red Sea [98]. However, the configuration of the amino acids in 160 has not been determined. Compound 160 inhibits the growth of human HeLa cervix carcinoma, murine CT26 colon carcinoma and human MM.1S multiple myeloma cell lines, with IC50 values of 11, 12 and 8 μg/mL, respectively. Investigation of Nocardiopsis lucentensis CNR-712 has led to the discovery of two new 3-methyl-4-ethylideneproline-containing (Leu/Trp-Pro-HomoArg) tripeptides, lucentamycins A and B (161 and 162), which show cytotoxicity toward the HCT-116 cell line, with IC50 values of 0.20 and 11 μM, respectively [99]. Two tyrosine-derived diketopiperazines, nocazines F (163) and G (164), have also been obtained by culture of Nocardiopsis sp. YIM M13066 [100]. Compound 163 is cytotoxic to H1299, HeLa, HL7702, MCF-7, PC3 and U251 cells, with IC50 values of 3.87, 4.47, 7.10, 3.86, 8.17 and 22.5 μM, respectively, and compound 164 shows cytotoxicity toward H1299, HeLa, HL7702, MCF-7 and PC3 cells, with IC50 values of 2.60, 3.97, 8.73, 6.67 and 16.7 μM, respectively. Two new peptaibols, microbacterins A (165) and B (166), have been obtained from Microbacterium sediminis sp. nov. YLB-01(T) [101]. Compound 165 is toxic toward HCT-8, Bel-7402, BGC-823, A549 and A2780 cells, with IC50 values of >10, 1.98, 2.11, 2.30 and >30 μM, respectively. Compound 166 displays cytotoxicity toward the same cell lines, with IC50 values of 5.93, 1.94, 1.03, 2.08 and 3.79 μM, respectively.

            Figure 5 |

            Structures of compounds 149–182.

            Streptomyces sp. CNQ-593, isolated from a sediment sample collected at a depth of approximately 20 m near the island of Guam, has yielded three hexadepsipeptides (AMDA-γOHPip-HAA-αMeSer-γOHPip-γClPip), piperazimycins A–C (167–169) [102]. Compounds 167–169 exhibit cytotoxicity toward HCT-116 cells, with the same GI50 value of 76 ng/mL. Compound 167 also displays significant cytotoxicity toward 60 tumor cell lines. One 15-membered depsipeptide, rakicidin D (170), has been isolated from Streptomyces sp. MWW064 from a marine sediment sample collected in Samut Sakhon province, Thailand [103]. Compound 170 contains an N-Me glycine moiety, a β-hydroxyasparagine moiety, 2,4-dimethyl-3-hydroxydecanoate moiety and γ-amino-2,4-pentadienoate moiety. However, the stereochemistry of compound 170 has not been investigated. Compound 170 shows cytotoxicity toward murine carcinoma colon 26-L5 cells, with an IC50 value of 6.7 μM. Two new highly modified linear tetrapeptides, padanamides A (171) and B (172), have been isolated from Streptomyces sp. RJA2928 [104]. Compound 171 is composed of a 2-methoxyacetic acid (Maa), 3-hydroxyleucine (Hleu), piperazic acid (Pip), 4-amino-3-hydroxy-2-methyl-5-phenylpentanoic acid (Ahmpp) and 3-amino-2-oxopyrrolidine-1-carboxamide (Aopc) residue. Compound 172 is almost the same as 171 except for an Apd in 172 instead of an Aopc in 171. Compounds 171 and 172 show weak antitumor activity toward Jurkat T lymphocyte cells (ATCC TIB-152), with IC50 values of 60 and 20 μg/mL, respectively. A new thiodepsipeptide, verrucosamide (173), has been isolated from Verrucosispora sp. CNX-026 [105]. Compound 173 is a cyclic octapeptide (cyclo-(Gly-Cys-Ala-Cys-Gly-Cys-Ala-Cys)) connected to two 3-hydroxy-quinaldic acid moieties. Compound 173 shows activity toward MDA-MB-468 breast carcinoma and COLO 205 colon adenocarcinoma cells, with LD50 values of 1.26 and 1.4 μM. Compound 173 displays moderate cytotoxicity toward NCI 60.

            Among these 27 non-ribosomal peptides and hybrids of polyketides and peptides (147–173), compounds 167–169 show potent cytotoxicity, with IC50 values at ng/mL (nM) levels. In the past few decades, scientists have been overcoming the well-known limitations of bioactive peptides as therapeutics. More peptides or peptide derivatives have been approved for clinical use. Hence, these three cyclic peptides (167–169), each with three piperazic acid units, are worthy of further investigation.

            2.3 Isoprenoids, terpenoids, sterols and hybrids of isoprenoids and peptides (or polyketides)

            A new sesquiterpene, 15-hydroxy-T-muurolol (174), produced by Streptomyces sp. M491, has been found to exhibit weak cytotoxic effects toward 37 human tumor cell lines, with a mean IC50 value of 6.7 μg/mL [106]. A new ergosterol, ananstrep C (175), has been isolated from Streptomyces anandii H41-59 and found to display cytotoxic activity toward SF-268, MCF-7 and NCI-H460 cells, with IC50 values of 13.0, 18.1 and 23.5 μg/mL, respectively [107]. The culture of Actinomadura sp. SBMs009 has afforded a 3-keto sterol compound, bendigole D (176), which shows cytotoxic activity toward L929 cells, with an IC50 value of 30 μM [108].

            Streptomyces sp. NBRC105896 has been found to produce a new teleocidin analog, JBIR-31 (177) [109]. Compound 177 is composed of a monoterpenoid moiety, N-methyl valine moiety and tryptophan moiety. Compound 177 displays cytotoxicity toward HeLa and ACC-MESO-1 cells, with IC50 values of 49 and 88 μM, respectively. Streptomyces sp. CHQ-64 has been found to produce drimentine I (178) [110]. Compound 178 is a hybrid of a sesquiterpenoid and a diketopiperazine (Val-Trp), possessing a rare heptacyclic skeleton. Compound 178 shows cytotoxicity toward HeLa cells, with an IC50 value of 16.73 μM.

            Streptomyces sp. CNQ-027 has afforded a new meroterpenoid, actinoranone (179), which is a hybrid of a diterpenoid and a polyketide. Compound 179 displays cytotoxicity toward the HCT-116 cell line, with an LD50 value of 2.0 μg/mL [111]. Three farnesyl-nitropyrroles—nitropyrrolin A (180), nitropyrrolin B (181) and nitropyrrolin D (182)—have been obtained from marine actinomycete strain CNQ-509 [112]. The structures of the nitropyrrolins (180–182) are composed of α-nitropyrroles with functionalized farnesyl groups at the C-4 position. Biogenetically, the pyrroles might be derived from succinyl-CoA and glycine. Compounds 180–182 exhibit cytotoxic activity toward HCT-116 cells, with IC50 values of 31.1, 31.0 and 5.7 μM, respectively. Streptomyces niveus SCSIO 3406 has been found to produce the new geranylated phenazines marfuraquinocins A−D (183–186; Figure 6 ) [113]. Compounds 183–186 each contain a sesquiterpenoid and a naphthoquinone moiety. Compounds 183–186 show cytotoxicity toward SF-268, MCF-7, NCI-H460 and HepG2 cells, with IC50 values in the range of 3.7 to 27.9 μM. Napyradiomycins A and D–F (187–190) have been obtained from liquid culture of Streptomyces sp. CNQ-329 [114]. Compounds 187–190 are each composed of a C5 isoprenoid moiety, monoterpenoid moiety and naphthoquinone moiety, and each shows cytotoxicity toward HCT-116 cells, with IC50 values of 4.19, 16.1, 4.81 and 9.42 μg/mL, respectively. Four napyradiomycin derivatives, napyradiomycin CNQ525.510B, napyradiomycin CNQ525.538, napyradiomycin CNQ525.554 and napyradiomycin CNQ525.600 (191–194), have been obtained from a culture of actinomycete strain CNQ525 [115]. Compounds 191, 192 and 194 have been found to be active toward HCT-116, with IC50 values of 17, 6 and 49 μM, respectively. A new pyrrolosesquiterpene, glaciapyrrole A (195), is produced by Streptomyces sp. NPS008187 [116]. Compound 195 displays inhibitory activity toward HT-29 and B16-F10 cells, with an IC50 value of 180 μM for both.

            Figure 6 |

            Structures of compounds 183–242.

            None of these 22 isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides) (174–195) demonstrate potent cytotoxicity. In addition, all the 22 structures were new at the time of their discovery. However, the linearly tricyclic/tetracyclic compounds 187 and 188 each uniquely have a C8/C6 linker bridging the first and second/third rings (C-2–C5a/C-2–C-6a).

            2.4 Heterocyclic, (hetero)aromatic and other compounds

            Two new 3,6-disubstituted indoles (196 and 197) have been obtained from Streptomyces sp. BL-49-58-005 [117]. Compound 196 shows cytotoxicity toward the K562 cell line, with a GI50 value of 8.46 μM. Compound 197 exhibits activity with GI50 values within the micromolar range against LN-caP, HMEC1, K-562, PANC1, LOVO and LOVO-DOX, and slightly higher values against other tumor cell lines, without any particular specificity. A 10H-phenoxazine derivative, strepoxazine A (198), has been identified from the solid culture of Streptomyces sp. SBT345 and found to exhibit cytotoxicity toward HL-60, with an IC50 of 8 μM [118]. Two pentacyclic indolosesquiterpenes, xiamycin (199) and its methyl ester (200), have been discovered from Streptomyces sp. GT2002/1503, an endophyte from the mangrove plant Bruguiera gymnorrhiza [119]. Compound 199 shows moderate cytotoxicity toward 12 tumor cell lines, whereas 200 is cytotoxic to 12 different tumor cell lines, with a geometric mean IC50 value of 10.4 μM.

            Streptomyces sioyaensis SA-1758 produces a 2,4a,5,6,7,7a-hexahydro-1H-cyclopenta[c]pyridine derivative, altemicidin (201), which inhibits L1210 and IMC carcinoma cells, with IC50 values of 0.84 and 0.82 μg/mL, respectively [120]. A unique phenyl benzoate derivative with an N-acetyl-(S)-cysteine moiety, bagremycin C (202), has been isolated from Streptomyces sp. Q22 and found to display cytotoxicity toward U87MG, U251, SHG44 and C6 cells, with IC50 values of 2.2, 4.3, 2.4 and 6.4 μM, respectively [121]. Streptomyces sp. KORDI-3238 has yielded streptokordin (203), 4-acetyl-6-methylpyridin-2(1H)-one [122]. Compound 203 exhibits cytotoxicity toward MDA-MB-231, HCT15, PC-3, NCI-H23, ACHN, LOX-IMVI and K-562 cells, with IC50 values of 7.5, 7.8, 3.2, 3.5, 4.7, 7.4 and 8.6 μg/mL, respectively. Two phenyl(1H-pyrrol-2-yl)methanone dimers, marinopyrroles A (204) and B (205), have been obtained from Streptomyces sp. CNQ-418, and 204 and 205 exhibit cytotoxic activity toward HCT-116, with IC50 values of 8.8 and 9.0 μM, respectively [123]. Although configurationally stable at room temperature, M-(–)-204 can be racemized at elevated temperatures, thus yielding the non-natural P-(+)-atropo-enantiomer, which inhibits HCT-116 with an IC50 value of 9.4 μM. The same strain, Streptomyces sp. CNQ-418, also produces marinopyrroles C–F (206–209) [124]. Compound 206 shows cytotoxic activity toward HCT-116, with an IC50 value of 0.21 μg/mL. Compounds 207–209 might be also active, but their cytotoxic activity has not been reported. Two novel spiroaminals, marineosins A and B (210 and 211), containing two pyrrole functionalities, have been isolated from cultures of the marine sediment-derived actinomycete Streptomyces sp. CNQ-617 [125]. Compounds 210 and 211 are active toward HCT-116 cell lines, with IC50 values of 0.5 and 46 μM, respectively. The difference in configuration at the spiroaminal center and in the tetrahydropyran conformation appears to significantly affect the bioactivity of compounds 210 and 211. An investigation of Streptomyces sp. CNR-698 has afforded ammosamides A (212) and B (213) [126]. The strain CNR-698 has been isolated from bottom sediments collected at a depth of 1,618 meters in the Bahamas. Compound 213 is a pyrrolo[4,3,2-de]quinolin-2(1H)-one derivative, whereas 212 is a thiol amide (-NH-CS-) instead of an amide (-NH-CO-), and has potential to exist in an equilibrium with its bis-iminoquinone tautomer. Compounds 212 and 213 are cytotoxic toward the HCT-116 cell line, with IC50 values of 320 nM for both. The isoquinolinequinone derivatives mansouramycins A–C (214–216) have been obtained from Streptomyces sp. isolate Mei37 [127]. Compound 216 has been demonstrated to be the most active compound, with an overall potency of 89 nM (mean IC50 value toward 36 tumor cell lines tested), followed by 215 (mean IC50 value 2.7 μM). Compound 214 shows moderate concentration-dependent cytotoxicity, with a mean IC50 value of 13.44 μM. N1-acetyl-N7-phenylacetyl cadaverine (217) has been discovered from Streptomyces sp. WuXin, and the bisamide shows activity toward HL-60 cells, with an IC50 value of 58.43 μM [128]. Streptomyces fradiae 007 M135 has afforded three new indolocarbazoles, fradcarbazoles A–C (218–220) [129]. Structurally, compounds 218–220 are very similar to staurosporine, with the N-Me replaced by various functional groups. Compounds 218–220 are strongly cytotoxic toward the HL-60/K562/A-549/BEL-7402 cell lines, with IC50 values of 1.30/4.58/1.41/3.26, 1.60/1.47/ 0.001/1.74 and 0.13/0.43/0.02/0.68 μM, respectively. Streptomyces sp. SCSIO 03032 has afforded three bisindole alkaloids, spiroindimicins B–D (221–223) [130]. Compounds 221–223 are unique molecules, each containing six rings. The cyclic pentane ring in the tetrahydrocyclopenta[c]pyrrole moiety is fused to one indole moiety and forms a spiro functionality with another indole moiety. Compound 221 displays cytotoxic activity against CCRF-CEM, B16 and H460 cells, with IC50 values of 4, 5 and 12 μg/mL, respectively. Compound 222 inhibits HepG2 and H460 cells, with IC50 values of 6 and 15 μg/mL, respectively. Compound 223 displays moderate cytotoxic activity toward HepG2, H460 and B16 cells. A new staurosporinone, N-carboxamido-staurosporine (224), has been isolated from the culture broth of the marine-derived Streptomyces sp. QD518 [131]. Biogenetically, 224 can be derived from two molecules of tryptophan and an amino sugar. Compound 224 exhibits cytotoxic activity toward 37 human tumor cell lines, with a mean IC50 value of 16 ng/mL. Streptomyces sp. SCSIO 03032 has produced a new bisindole alkaloid, indimicin B (225), a compound similar to staurosporine without the amino sugar moiety [132]. Compound 225 exhibits cytotoxic activity toward MCF-7 cells, with an IC50 value of 10.0 μM. Investigation of Streptomyces variabilis SNA-020 has led to the isolation of a quinoline-5,6-dione derivative, ammosamide D (226) [133]. Strain SNA-020 has been isolated from a sediment sample collected at Sweetings Cay, Bahamas. Compound 226 displays cytotoxic activity toward MIA PaCa-2 cells, with an IC50 value of 3.2 μM. Streptocarbazoles A and B (227 and 228) have been isolated from Streptomyces sp. FMA [134]. Compounds 227 and 228 are staurosporine analogs with differences at C-3–C-5 in the amino sugar moiety. Compound 227 shows cytotoxicity toward HL-60, A549, P388 and Hela cell lines, with IC50 values of 1.4, 5.0, 18.9 and 34.5 μM, respectively. Compound 228 is active toward P388 and Hela cell lines, with IC50 values of 12.8 and 22.5 μM, respectively. An aminophenoxazinone alkaloid, maroxazinone (229), has been discovered from Streptomyces sp. Eg25, and shown activity against MCF-7, HEPG-2 and HCT-116 cells, with IC50 values of 4.32, 2.90 and 8.51 μg/mL, respectively [135]. Streptomyces niveus SCSIO 3406 has produced two new geranylated phenazines, phenaziterpene A (230) and phenaziterpene B (231). [113] Compounds 230 and 231 are hybrids of a monoterpenoid and a phenazine moiety, probably derived from chorismic acid. Both 230 and 231 show cytotoxicity toward SF-268, MCF-7 and HepG2 cells, with IC50 values ranging from 10.2 to 52.7 μM, and 230 is weakly active toward NCI-H460, with an IC50 value of 68.9 μM. Streptomyces sp. CNS284 has afforded two phenazines (232 and 233), which induce apoptosis in HL-60 cells [136]. Two new hexahydro-1H-pyrrolizine dimers, dibohemamines B and C (234 and 235), have been isolated from an extract of a cultured marine-derived Streptomyces spinoverrucosus SNB-032 [137]. Compounds 234 and 235 exhibit cytotoxic activity toward the A549 cell line, with IC50 values of 0.140 and 0.145 μM, respectively. Compounds 234 and 235 also show inhibitory activity toward HCC1171 cells, with IC50 values of 3.9 and 1.2 μM, respectively. In addition, compounds 234 and 235 inhibit HCC44 and HCC366 cells, respectively, with IC50 values of 12.0 and 6.7 μM, respectively. An unique molecule composed of a pyrrolo[2,1-a]isoindole and a pyrrolizine moieties, chlorizidine A (236), has been isolated from Streptomyces sp. CNH-287 and found to inhibit HCT-116 cells, with an IC50 value of 3.2–4.9 μg/mL [138]. Four new cyclic bipyridine glycosides, cyanogrisides E–H (237–240), have been isolated from Actinoalloteichus cyanogriseus WH1-2216-6 [139]. Compounds 237 and 240 show cytotoxicity toward K562 cells, with IC50 values of 6.0 and 0.8 μM, respectively. Compounds 238 and 239 inhibit A549, K562, HeLa, HCT116 and HL-60 cells, with IC50 values of 33.1/42/0. 13.6/23/6, 26.5/44.1, 0.8/3.6 and 3.1/2.0 μM, respectively. Saccharomonosporine A (241) has been isolated from the extract of Saccharomonospora sp. UR22 and Dietzia sp. UR66 co-culture [140]. Compound 241 is a brominated oxo-indole alkaloid connected to a 4-methoxy benzene ring through an imine containing linker (=CH-C(=NH)-CH=CH-). Compound 241 shows cytotoxicity toward the cancer cell lines HT-29 and HL-60, with IC50 values of 3.6 and 2.8 μM, respectively. Two prodiginine derivatives, cyclononylprodigiosin (242) and nonylprodigiosin (243; Figure 7 ), have been obtained from the actinomycete strain BRA 177 [141]. Compounds 242 and 243 are cytotoxic toward SK-Mel-147, HCT-116 and MRC-5 cells, with IC50 values of 2.40/2.70, 3.94/4.25 and 0.58/0.26 μM, respectively.

            Figure 7 |

            Structures of compounds 243254.

            Amycolactam (244), an indole alkaloid with an isoprenyl group at C-4 and 5-methyl-pyrrolidin-2-one moiety at C-3, has been discovered from the sponge-associated rare actinomycete Amycolatopsis sp., and found to cytotoxic to SNU638 and HCT116 cells, with IC50 values of 0.8 and 2.0 μM, respectively. Compound 244 is also cytotoxic to A546, K562 and SK-HEP1 cells, with IC50 values of 13.7, 9.6 and 8.3 μM, respectively [142]. Ten 2,9-dihydro-1H-pyrido[3,4-b]indol-1-one alkaloids—marinacarbolines E–G (245–247), I (248), K–M (249–251), O (252), Q (253) and caerulomycin N (254)—have been obtained from the actinomycete strain Actinoalloteichus sp. ZZ1866 [143]. Compounds 245–254 exhibit cytotoxic activity toward U87MG and U251 cells, with IC50 values in the range of 2.0–43 μM.

            Among these 59 compounds classified as heterocyclic, (hetero)aromatic and other (196–254), compounds 212, 213, 216 and 224 show potent cytotoxicity, with IC50 values at the ng/mL (nM) level. Structurally, most of these 59 compounds markedly differ from the 195 compounds in Sections 2.1, 2.2 and 2.3, because of their heterocyclic or (hetero)aromatic moieties.

            3. CONCLUSION

            From 1989 until the end of 2020, 254 new cytotoxic compounds have been obtained from marine actinomycetes. This review summarized the structures, strain sources, and cytotoxicity of these secondary metabolites ( Table 1 ). Most of the compounds (206) were reported from 2010 to 2020 ( Figure 8 ). The numbers of newly reported compounds have increased since 1989, peaked in the mid-2010s (2013–2017) and decreased in the following years. However, we expect the numbers to increase after the COVID-19 pandemic ends. Of these 254 compounds, most are moderately active, but approximately 20 compounds show potent cytotoxicity with IC50 values at the ng/mL/nM level (see the Prospects section). The articles reporting these compounds have been published in 30 different journals, and the “Journal of Natural Products” (72) published more articles than any other single journal, followed by “Marine Drugs” (36), “Organic Letters” (27), the Journal of Antibiotics” (21), and the “Journal of Organic Chemistry” (18; Figure 9 ). Interestingly, beyond these prominent natural-product journals, “Phytochemistry” published seven articles, although it is a peer-reviewed scientific journal covering pure and applied plant chemistry, plant biochemistry and molecular biology. This review classified the compounds into four classes: polyketides; non-ribosomal peptides, and hybrids of polyketides and peptides; isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides); and heterocyclic, (hetero)aromatic and other compounds. These cytotoxic compounds have diverse chemical structures, and most are polyketides (146) making up 58% of the 254 new antitumor compounds ( Figure 10 ). Among these 146 polyketides, most are categorized as either macrolides (lactones), lactams and α/γ-pyrones (57), or benzoquinones, naphthoquinones, anthraquinones and other aromatic compounds (57), which together accounted for 45% of the total 254 compounds.

            Table 1 |

            Cytotoxic compounds isolated from marine actinomycetes.

            CompoundProducing strainStrain sourceArchitectural featureReferences
            1–2 Streptomyces carnosus AZS17Coastal waters of the East China SeaPolyketides[11]
            3 Streptomyces sp. PG-19Surface of the Sea of Cortez gorgonian octocoral Pacifigorgia sp.Polyketides[12]
            4–5 Streptomyces caniferus GUA-06-05-006AMarine-derived culture brothPolyketides[13]
            6 Streptomyces sp. GIC10-1Marine sponge collected off the coast of Kenting, TaiwanPolyketides[14]
            7–8 Streptomyces sp. GIC10-1Bacterial communities associated with the marine sponge Theonella sp.Polyketides[15]
            9 Streptomyces sp. M-207Cold-water coral Lophelia pertusa Polyketides[16]
            10 Streptomyces sp. SCSIO 01127South China Sea sedimentPolyketides[17]
            11–12 Streptomyces sp.Crown-of-thorns starfish, Acanthaster planci Polyketides[18]
            13 Nocardiopsis sp. NHF48South China Sea sedimentsPolyketides[19]
            14–17 Streptomyces sp. HKI0576Polyketides[20]
            18 Streptomyces aureoverticillatus NPS001583Marine sedimentPolyketides[21]
            19–20 Streptomyces sp. HZP-2216ETraditional Chinese medicine sea lettuce Ulva pertusa (family Ulvaceae)Polyketides[22]
            21 Streptomyces althioticus MSM3Intertidal seaweed Ulva sp., Cantabrian Sea (Northeast Atlantic Ocean)Polyketides[23]
            22 Streptomyces sp. 219807Mangrove soil from SanyaPolyketides[24]
            23 Streptomyces sp. ART5Surface sediment from the East Siberian continental marginPolyketides[25]
            24–27 Streptomyces sp. THS-55Conserved mangrove in Hainan province, ChinaPolyketides[26]
            28–29 Streptomyces antibioticus H12-15Sea sediment from a mangrove in the South China SeaPolyketides[27]
            30–32 Streptomyces sp. DSS-18Deep-sea sediment from the West PacificPolyketides[28]
            33 Streptomyces sp. MDG-04-17-069Marine sediment from the east coast of Madagascar, 30 m depthPolyketides[29]
            34–35 Streptomyces sp. 211726Mangrove rhizosphere soilPolyketides[30]
            36–42 Streptomyces sp. 211726Mangrove brothPolyketides[31]
            43–48 Streptomyces pactum SCSIO02999Polyketides[32]
            49–50 Streptomyces sp.Estuary between the Yellow Sea and the Han River, Republic of KoreaPolyketides[33]
            51 Pseudonocardia sp. HS7Cloacal aperture of the sea cucumber Holothuria moebii Polyketides[34]
            52–54 Nocardiopsis CG3 (DSM 106572)Saltpan of KenadsaPolyketides[35]
            55 Micromonospora sp. FIM05328Soil sample from the East China SeaPolyketides[36]
            56 Micromonospora strain FIM07-0019Shallow coastal waters near the island of Chiloe, ChilePolyketides[37]
            57 Actinoalloteichus cyanogriseus WH1-2216-6Submarine sedimentPolyketides[38]
            58 Streptomyces sp. Sp080513GE- 26 Haliclona sp. marine spongePolyketides[39]
            59 Streptomyces sp. SpD081030ME-02Demospongiae class of marine sponge, offshore of Ishigaki City, Okinawa Prefecture, JapanPolyketides[40]
            60 Streptomyces sp. HB202 Halichondria panicea spongePolyketides[41]
            61–63 Streptomyces sp. BCC45596ThailandPolyketides[42]
            64–66 Streptomyces sp. M7_15Caribbean spongesPolyketides[43]
            67–68 Streptomyces sp. OUCMDZ-1703Soft coralPolyketides[44]
            69 Streptomyces sp. HDN-10-293SpongePolyketides[45]
            70 Streptomyces sp. RKBHB7 Eunicea sp. unidentified octocoralPolyketides[46]
            71–72 Streptomyces sp. ZZ406 Haliplanella lineata sea anemonePolyketides[47]
            73–76 Streptomyces sp. (CANU Fox 21-2-6)New Zealand micro-organismsPolyketides[48]
            77 Streptomyces sp. FX-58 Salicornia herbacea Polyketides[49]
            78 Streptomyces sp. B8652Polyketides[50]
            79–80 Streptomyces sp. M045Polyketides[51]
            81–82 Streptomyces sp. CNH990Marine sedimentsPolyketides[52]
            83–85 Streptomyces sp. NPS853Marine sedimentsPolyketides[53]
            86 Streptomyces sp. W007Polyketides[54]
            87–90 Streptomyces lusitanus SCSIO LR32Marine sediments from South China SeaPolyketides[55]
            91–92 Streptomyces sp. (strain SNE-011)Sediment sample from Kiawah Island, South Carolina,Polyketides[56]
            93 Streptomyces H74-21Sea sediment in a mangrove sitePolyketides[57]
            94 Streptomyces sp. SCSIO 11594South China Sea sediment, 2,403 m depthPolyketides[58]
            95–96 Streptomyces sp. IFM11490Soil and seawater samples from different areas of Japan.Polyketides[59]
            97–98 Streptomyces sp. 182SMLYMarine sedimentsPolyketides[60]
            99 Streptomyces sp. F001Polyketides[61]
            100 Streptomyces griseus M268Sediment from Kiaochow Bay, ChinaPolyketides[62]
            101 Micromonospora lomaivitiensis LL-37I366Polyketides[63]
            102–103 Streptomyces fradiae 007Polyketides[64]
            104 Streptomyces lusitanus SCSIO LR32Deep seaPolyketides[65]
            105–106 Streptomyces lusitanus SCSIO LR32Deep seaPolyketides[66]
            107–108 Streptomyces sp. XMA39Polyketides[67]
            109 Micromonospora sp. G039Sediment from the Cát Bà peninsula, East Sea of VietnamPolyketides[68]
            110–112 Micromonospora echinospora SCSIO 04089Sediment from the northern South China Sea, 3,025 m depthPolyketides[69]
            113 Saccharothrix sp. 10-10Polyketides[70]
            114 Nonomuraea sp. AKA32Deep-sea water from Sagami Bay, Japan, 800 m depthPolyketides[71]
            115 Streptomyces sp. RJA2928Polyketides[72]
            116–119 Streptomyces sp. RJA2928Polyketides[73]
            120 Streptomyces sp. 0616208Polyketides[74]
            121–122 Streptomyces sp. YM14-060Polyketides[75]
            123 Streptomyces nodosus NPS007994Marine sediment from Scripps Canyon, La Jolla, CaliforniaPolyketides[76]
            124 Streptomyces sp. CHQ-64Polyketides[77]
            125–128 Streptomyces albus POR-04-15-053Extracts of the air-breathing gastropod Siphonaria diemensis, a marine molluskPolyketides[78]
            129 Streptomyces sp. B8112Polyketides[79]
            130 Streptomyces sp. TP-A0879Stem of the bracken Pteridium aquilinum Polyketides[80]
            131 Streptomyces sp. SAK1Southern area of Jeju Island, Republic of KoreaPolyketides[81]
            132 Streptomyces violans HTTA-F04129 Saliconia sp. from the intertidal zone of Rushan County, Shandong PeninsulaPolyketides[82]
            133 Streptomyces sp. SNJ210Deep-sea areas from Jeju Island, KoreaPolyketides[83]
            134–137 Streptomyces sp. CNQ-085Polyketides[84]
            138–139 Streptomyces sp. NPS-643Marine sediment sample near Kochi Harbor, Japan, 30 m depthPolyketides[85]
            140–141 Streptomyces sp. HNA39Polyketides[86]
            142 Micromonospora sp. 29867Suruga Bay, Shizuoka Prefecture, JapanPolyketides[87]
            143 Micromonospora matsumotoense M-412Cantabrian Sea sediments, 2,000 m depthPolyketides[88]
            144–146 Verrucosispora sp. SCSIO 07399Deep-sea marine sedimentPolyketides[89]
            147 Streptomyces sp. LS298 Gelliodes carnosa marine sponge from the South China SeaNon-ribosomal peptides, and hybrids of polyketides and peptides[90]
            148 Streptomyces sp. SBT348 Petrosia ficiformis Mediterranean sponge from Milos, GreeceNon-ribosomal peptides, and hybrids of polyketides and peptides[91]
            149 Streptomyces sp. SNJ013Deep-sea sediment from Jeju Island, KoreaNon-ribosomal peptides, and hybrids of polyketides and peptides[92]
            150–151 Streptomyces sp. SNJ042Sand beach at Jeju, a volcanic island in the Republic of KoreaNon-ribosomal peptides, and hybrids of polyketides and peptides[93]
            152–153 Streptomyces sp. P11-23BNon-ribosomal peptides, and hybrids of polyketides and peptides[94]
            154 Streptomyces sp. SSA 13Arabian Sea sediments from the eastern edge of the seashoreNon-ribosomal peptides, and hybrids of polyketides and peptides[95]
            155–157 Streptomyces sp. MNU FJ-36Intestinal fabric of Katsuwonus sp.Non-ribosomal peptides, and hybrids of polyketides and peptides[96]
            158–159 Streptomyces sp. Did-27Marine microbial bioactive leadsNon-ribosomal peptides, and hybrids of polyketides and peptides[97]
            160 Nocardiopsis sp. UR67Red SeaNon-ribosomal peptides, and hybrids of polyketides and peptides[98]
            161–162 Nocardiopsis lucentensis (strain CNR-712)Sediment from a shallow saline pond on the island of Little San Salvador, Bahamas.Non-ribosomal peptides, and hybrids of polyketides and peptides[99]
            163–164 Nocardiopsis sp. YIM M13066Deep-sea sedimentNon-ribosomal peptides, and hybrids of polyketides and peptides[100]
            165–166 Microbacterium sediminis sp. nov. YLB-01(T)Deep seaNon-ribosomal peptides, and hybrids of polyketides and peptides[101]
            167–169 Streptomyces sp. CNQ-593Marine sediments near the island of GuamNon-ribosomal peptides, and hybrids of polyketides and peptides[102]
            170 Streptomyces sp. MWW064Marine sediment from Samut Sakhon province, ThailandNon-ribosomal peptides, and hybrids of polyketides and peptides[103]
            171–172 Streptomyces sp. RJA2928Crude organic extracts from marine sediment collected near the passage Padana Nahua, Papua New GuineaNon-ribosomal peptides, and hybrids of polyketides and peptides[104]
            173 Verrucosispora sp. CNX-026Non-ribosomal peptides, and hybrids of polyketides and peptides[105]
            174 Streptomyces sp. M491Sand sample from Qingdao (China)Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[106]
            175 Streptomyces anandii H41-59Sea sediment from a mangrove districtIsoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[107]
            176 Actinomadura sp. SBMs009New marine spongeIsoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[108]
            177 Streptomyces sp. NBRC105896 Haliclona sp.Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[109]
            178 Streptomyces sp. CHQ-64Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[110]
            179 Streptomyces sp. CNQ-027Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[111]
            180–182 Actinomycete family Streptomycetaceae CNQ-509Marine sediment from La Jolla, CaliforniaIsoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[112]
            183–186 Streptomyces niveus SCSIO 3406South China Sea sediment, 3,536 m depthIsoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[113]
            187–190 Streptomyces sp. CNQ-329Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[114]
            191–194 Actinomycete strain CNQ525Isoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[115]
            195 Streptomyces sp. NPS008187Marine sediment from AlaskaIsoprenoids, terpenoids, sterols, and hybrids of isoprenoids and peptides (or polyketides)[116]
            196–197 Streptomyces sp. BL-49-58-005Unidentified marine invertebrate from MexicoHeterocyclic, (hetero)aromatic and other compounds[117]
            198 Streptomyces sp. SBT345 Agelas oroides Mediterranean spongeHeterocyclic, (hetero)aromatic and other compounds[118]
            199–200 Streptomyces sp. GT2002/1503Stem of Bruguiera gymnorrhiza Heterocyclic, (hetero)aromatic and other compounds[119]
            201 Streptomyces sioyaensis SA-1758Sea mud from Gamo, Miyagi Prefecture, JapanHeterocyclic, (hetero)aromatic and other compounds[120]
            202 Streptomyces sp. Q22Mangrove soilHeterocyclic, (hetero)aromatic and other compounds[121]
            203 Streptomyces sp. KORDI-3238Deep-sea sedimentHeterocyclic, (hetero)aromatic and other compounds[122]
            204–205 Streptomyces sp. CNQ-418Marine sediment from La Jolla, California, 51 m depthHeterocyclic, (hetero)aromatic and other compounds[123]
            206–209 Streptomyces sp. CNQ-418Marine sediment from La Jolla, California, 51 m depthHeterocyclic, (hetero)aromatic and other compounds[124]
            210–211 Streptomyces sp. CNQ-617Heterocyclic, (hetero)aromatic and other compounds[125]
            212–213 Streptomyces sp. CNR-698BahamasHeterocyclic, (hetero)aromatic and other compounds[126]
            214–216 Streptomyces sp. Mei37Muddy sediment from Jade Bay, southern German North Sea coastHeterocyclic, (hetero)aromatic and other compounds[127]
            217 Streptomyces sp. WuXinHeterocyclic, (hetero)aromatic and other compounds[128]
            218–220 Streptomyces fradiae 007M135Sediment from Jiaozhou Bay, Shandong Province, ChinaHeterocyclic, (hetero)aromatic and other compounds[129]
            221–223 Streptomyces sp. SCSIO 03032Deep-sea sedimentHeterocyclic, (hetero)aromatic and other compounds[130]
            224 Streptomyces sp. QD518Heterocyclic, (hetero)aromatic and other compounds[131]
            225 Streptomyces sp. SCSIO 03032Deep-sea sedimentHeterocyclic, (hetero)aromatic and other compounds[132]
            226 Streptomyces variabilis SNA-020Heterocyclic, (hetero)aromatic and other compounds[133]
            227–228 Streptomyces sp. FMAMangrove soil from Sanya, Hainan province, ChinaHeterocyclic, (hetero)aromatic and other compounds[134]
            229 Streptomyces sp. Eg25Heterocyclic, (hetero)aromatic and other compounds[135]
            230–231 Streptomyces niveus SCSIO 3406South China Sea sediment, 3,536 m depthHeterocyclic, (hetero)aromatic and other compounds[113]
            232–233 Streptomyces sp. CNS284Heterocyclic, (hetero)aromatic and other compounds[136]
            234–235 Streptomyces spinoverrucosus strain SNB-032Heterocyclic, (hetero)aromatic and other compounds[137]
            236 Streptomyces sp. strain CNH-287Heterocyclic, (hetero)aromatic and other compounds[138]
            237–240 Actinoalloteichus cyanogriseus WH1-2216-6Heterocyclic, (hetero)aromatic and other compounds[139]
            241 Saccharomonospora sp. UR22 and Dietzia sp. UR66Red Sea sponge Callyspongia siphonella Heterocyclic, (hetero)aromatic and other compounds[140]
            242–243 Actinomycete strain BRA 177Saint Peter and Saint Paul Archipelago, BrazilHeterocyclic, (hetero)aromatic and other compounds[141]
            244 Amycolatopsis sp.SpongeHeterocyclic, (hetero)aromatic and other compounds[142]
            245–254 Actinoalloteichus sp. ZZ1866Sea mud from the coastal area of Putuo, Zhoushan, ChinaHeterocyclic, (hetero)aromatic and other compounds[143]
            Figure 8 |

            Numbers of antitumor compounds isolated from marine actinomycetes each year (1989 to 2020).

            Figure 9 |

            Journals publishing, and numbers of articles describing, antitumor compounds from marine actinomycetes.

            Figure 10 |

            Structural classes of antitumor compounds from marine actinomycetes.

            Marine actinomycetes produce different biologically active secondary metabolites. In 2012, Subramani and Aalbersberg published an article in “Microbiological Research” indicating that marine actinomycetes are an ongoing source of novel bioactive metabolites [144]. In 2009, a review article reported antitumor compounds from marine actinomycetes [145]. In 2020 and 2021, we reported the sources of marine actinomycetes, chemical structures and biological activities of 127 halogenated compounds and 313 antimicrobial compounds from multiple marine actinomycetes [146, 147]. Marine actinomycetes are a promising source of lead compounds for drug discovery.

            Despite the discovery of many cytotoxic compounds from marine actinomycetes, several drawbacks of natural product anticancer drug discovery exist. Some cytotoxic compounds have been obtained through assay-guided separation, but in many cases, no assay-guided separation was performed, and cytotoxic compounds were identified simply through purification followed by cytotoxic evaluation. Most of the cytotoxic compounds have not been tested for their selectivity toward different cancer cell lines and normal human cell lines, mainly because of insufficient financial support to researchers. Bioassay-guided separation is sometimes very tedious, and dereplication does not always work well, as researchers expect. Because naturally occurring compounds in their original forms may not always be patentable in the USA, although simple derivatives can be patent protected, natural product chemists’ enthusiasm for anticancer drug discovery from natural sources has been diminished.

            Selection of strains, culturing strategies and analytical techniques for natural-product-library establishment and natural-product dereplication will be of great help in anticancer drug discovery from marine actinomycetes. A future direction may involve advancing genome mining and gene manipulation, as discussed below.

            4. PROSPECTS

            Some of the reviewed compounds have demonstrated potent cytotoxic activity, with IC50 values at ng/mL or nM levels, for example, compounds 3 [12], 6–8 [14], 55 [38], 73–76 [49], 81 [53], 101 [63], 121 and 122 [75], 124 [77], 167–169 [102], 212 and 213 [126], 216 [127] and 224 [131]. However, the selectivity of some potent cytotoxic compounds has not been investigated. Selectivity study is important, because identifying cytotoxic drugs with a high selectivity toward cancer cells is critical to increase the low survival rates of patients with cancer. One approach to avoiding adverse effects of cytotoxic agents is targeted drug delivery. For instance, a cytotoxic drug can be hung on an antibody scaffold to form an antibody–drug conjugate. Subsequently, the complex targeted agent can overcome the unspecific toxic effects of conventional drug delivery, thereby decreasing the amount of drug required for therapeutic efficacy.

            Most of the secondary metabolites reviewed herein have been evaluated for their antimicrobial and cytotoxic activities. Other biological properties could be identified through testing of actinomycete secondary metabolites in other biological settings. For example, the fungal metabolites sinuxylamides A and B have shown no antibacterial activity or cytotoxicity at 40 μM, but when tested for their antithrombotic activity, have demonstrated strong inhibition of the binding of fibrinogen to purified integrin IIIb/IIa in a dose-dependent manner, with IC50 values of 0.89 and 0.61 μM, respectively [148].

            Novel molecules with unprecedented structural and/or functional attributes usually have unique bioactivities. Some of the reviewed compounds in this article have unique structures, for example, compounds 1, 2, 9, 10, 23, 101, 123, 140, 141, 144–146 and most of the compounds classified as heterocyclic, (hetero)aromatic and other compounds (196–254). Compounds 101, 212, 213 and 224 are not only structurally interesting (particularly 101) but also exhibit potent cytotoxicity. The cytotoxicity of 101 arises from the induction of double-strand breaks in DNA [149]. Compound 101 has a molecular formula of C38H26N4O14. Its molecular weight is 762 Daltons, and the numbers of hydrogen-bond donors and acceptors in the molecule clearly violate Lipinski’s role of five; these findings, together with the compound’s structural complexity, suggest low druggability of 101. However, structural modification and/or formulation have made many undruggable compounds druggable. For example, halichondrin B (molecular formula: C60H86O19; molecular weight: 1111 Daltons) is a complex polyether macrolide originally isolated from the marine sponge Halichondria okadai [150], which was believed to be undruggable by many researchers. However, Eisai Co. has structurally simplified halichondrin B, and eribulin (brand name Halaven) was approved by the U.S. Food and Drug Administration on November 15, 2010, with an indication to treat metastatic breast cancer [151].

            Most of these 254 compounds are analogs of previously reported molecules. In general, structurally unique compounds represent a decreasing percentage of the total number of compounds isolated from natural sources in the past few decades. However, exploring unexplored and unusual source organisms, or those from unique environments, could provide opportunities for finding novel natural products.

            Currently, the genomes of actinomycete strains are routinely sequenced, and a host of bioinformatics tools are increasingly available for identifying potential biosynthetic gene clusters of actinomycete natural products. Developing universal expression systems for small-molecule biosynthesis with high yield, constructing genetic tools to access the biosynthetic potential of cultured marine actinomycetes and awakening “silent” biosynthetic pathways will be important approaches for discovery of small molecules from marine actinomycetes. Investigations aimed at understanding how the biosynthetic pathways operate at the genetic and biochemical levels in marine actinomycetes will open new doors to designing molecules with improved anticancer properties.

            ACKNOWLEDGEMENTS

            This work was supported in part by the Specific Research Project of Guangxi for Research Bases and Talents (AD 20297036); Guangxi Natural Science Foundation under grant number 2019GXNSFBA185002; and NIH NIGMS grant 5P20GM103466.

            CONFLICTS OF INTEREST

            The authors declare no conflicts of interest.

            REFERENCES

            1. Cappello E, Nieri P. From Life in the Sea to the Clinic: The Marine Drugs Approved and Under Clinical Trial. Life (Basel). 2021. Vol. 11:1390

            2. Carroll AR, Copp BR, Davis RA, Keyzers RA, Prinsep MR. Marine Natural Products. Natural Product Reports. 2022. Vol. 39:1122–1171

            3. Pye R, Bertin MJ, Lokey RS, Gerwick WH, Linington RG. Retrospective Analysis of Natural Products Provides Insights for Future Discovery Trends. Proceedings of the National Academy of Sciences. 2017. Vol. 114:5601–5606

            4. The marine pharmacology/pharmaceutical pipeline website. https://www.marinepharmacology.org/approvedAccessed on date July 13, 2022

            5. Gerwick WH, Moore BS. Lessons from the Past and Charting the Future of Marine Natural Products Drug Discovery and Chemical Biology. Chemistry & Biology. 2012. Vol. 19:85–98

            6. van der Meij A, Worsley SF, Hutchings MI, van Wezel GP. Chemical Ecology of Antibiotic Production by Actinomycetes. FEMS Microbiology Reviews. 2017. Vol. 41:392–416

            7. Donald L, Pipite A, Subramani R, Owen J, Keyzers RA, Taufa T. Streptomyces: Still the Biggest Producer of New Natural Secondary Metabolites, A Current Perspective. Microbiology Research. 2022. Vol. 13:418–465

            8. Joubert N, Beck A, Dumontet C, Denevault-Sabourin C. Antibody-Drug Conjugates: The Last Decade. Pharmaceuticals (Basel). 2020. Vol. 13:245

            9. Lamb YN. Inotuzumab Ozogamicin: First Global Approval. Drugs. 2017. Vol. 77:1603–1610

            10. Demain AL, Sanchez S. Microbial Drug Discovery: 80 Years of Progress. The Journal of Antibiotics. 2009. Vol. 62:5–16

            11. Wei RB, Xi T, Li J, Wang P, Li FC, Lin YC, et al.. Lobophorin C and D, New Kijanimicin Derivatives from a Marine Sponge-Associated Actinomycetal Strain AZS17. Marine Drugs. 2011. Vol. 9:359–368

            12. Tapiolas DM, Roman M, Fenical W, Stout TJ, Clardy J. Octalactins A and B: Cytotoxic Eight-Membered-Ring Lactones from a Marine Bacterium, Streptomyces sp. Journal of the American Chemical Society. 1991. Vol. 113:4682–4683

            13. Pérez M, Schleissner C, Fernández R, Rodríguez P, Reyes F, Zuñiga P, et al.. PM100117 and PM100118, New Antitumor Macrolides Produced by a Marine Streptomyces caniferus GUA-06-05-006A. The Journal of Antibiotics. 2016. Vol. 69:388–394

            14. Chen YH, Lu MC, Chung HM, Weng CF, Su JH, Yang YT, et al.. Bafilomycin M, a New Cytotoxic Bafilomycin Produced by a Streptomyces sp. Isolated from a Marine Sponge Theonella sp. Tetrahedron Letters. 2016. Vol. 57:4863–4865

            15. Chen YH, Yang JC, Lu MC, Weng CF, Su YD, Kuo J, et al.. Bafilomycins N and O, Novel Cytotoxic Bafilomycin Analogues Produced by Streptomyces sp. GIC10-1 Isolated from Marine Sponge Theonella sp. Tetrahedron. 2017. Vol. 73:5170–5175

            16. Braña AF, Sarmiento-Vizcaíno A, Osset M, Pérez-Victoria I, Martín J, Pedro ND, et al.. Lobophorin K, A New Natural Product with Cytotoxic Activity Produced by Streptomyces sp. M-207 Associated with the Deep-Sea Coral Lophelia pertusa . Marine Drugs. 2017. Vol. 15:144

            17. Niu S, Li S, Chen Y, Tian X, Zhang H, Zhang G, et al.. Lobophorins E and F, New Spirotetronate Antibiotics from a South China Sea-Derived Streptomyces sp. SCSIO 01127. Journal of Antibiotics. 2011. Vol. 64:711–716

            18. Shin HJ, Lee HS, Lee JS, Shin J, Lee MA, Lee HS, et al.. Violapyrones H and I, New Cytotoxic Compounds Isolated from Streptomyces sp. Associated with the Marine Starfish Acanthaster planci . Marine Drugs. 2014. Vol. 12:3283–3291

            19. Yang N, Song F. Bioprospecting of Novel and Bioactive Compounds from Marine Actinomycetes Isolated from South China Sea Sediments. Current Microbiology. 2018. Vol. 75:142–149

            20. Ding L, Maier A, Fiebig HH, Görls H, Lin WH, Peschel G, et al.. Divergolides A-D from a Mangrove Endophyte Reveal an Unparalleled Plasticity in Ansa-Macrolide Biosynthesis. Angewandte Chemie International Edition. 2011. Vol. 50:1630–1634

            21. Mitchell SS, Nicholson B, Teisan S, Lam KS, Potts BCM. Aureoverticillactam, a Novel 22-Atom Macrocyclic Lactam from the Marine Actinomycete Streptomyces aureoverticillatus . Journal of Natural Products. 2004. Vol. 67:1400–1402

            22. Zhang X, Chen L, Chai W, Lian XY, Zhang Z. A Unique Indolizinium Alkaloid Streptopertusacin A and Bioactive Bafilomycins from Marine-Derived Streptomyces sp. HZP-2216E. Phytochemistry. 2017. Vol. 144:119–126

            23. Braña AF, Sarmiento-Vizcaíno A, Pérez-Victoria I, Martín J, Otero L, Palacios-Gutiérrez JJ, et al.. Desertomycin G, a New Antibiotic with Activity Against Mycobacterium Tuberculosis and Human Breast Tumor Cell Lines Produced by Streptomyces althioticus MSM3, Isolated from the Cantabrian Sea Intertidal Macroalgae Ulva sp. Marine Drugs. 2019. Vol. 17:114–123

            24. Han Y, Tian E, Xu D, Ma M, Deng Z, Hong K. Halichoblelide D, a New Elaiophylin Derivative with Potent Cytotoxic Activity from Mangrove-Derived Streptomyces sp. 219807. Molecules. 2016. Vol. 21:970–976

            25. Moon K, Ahn CH, Shin Y, Won TH, Ko K, Lee SK, et al.. New Benzoxazine Secondary Metabolites from an Arctic Actinomycete. Marine Drugs. 2014. Vol. 12:2526–2538

            26. Zhang W, Che Q, Tan H, Qi X, Li J, Li D, et al.. Marine Streptomyces sp. Derived Antimycin Analogues Suppress HeLa Cells via Depletion HPV E6/E7 Mediated by ROS-dependent Ubiquitin-Proteasome System. Scientific Reports. 2017. Vol. 7:1–14

            27. Hu C, Zhou SW, Chen F, Zheng XH, Shen HF, Lin BR, et al.. Neoantimycins A and B, Two Unusual Benzamido Nine-Membered Dilactones from Marine-Derived Streptomyces Antibioticus H12-15. Molecules. 2017. Vol. 22:557–566

            28. Li J, Lu CH, Zhao BB, Zheng ZH, Shen YM. Phaeochromycins F–H, Three New Polyketide Metabolites from Streptomyces sp. DSS-18. Beilstein Journal of Organic Chemistry. 2008. Vol. 4:46

            29. Pérez M, Crespo C, Schleissner C, Rodríguez P, Zuñiga P, Reyes F. Tartrolon D, a Cytotoxic Macrodiolide from the Marine-Derived Actinomycete Streptomyces sp. MDG-04-17-069. Journal of Natural Products. 2009. Vol. 72:2192–2194

            30. Yuan G, Lin H, Wang C, Hong K, Liu Y, Li J. 1H and 13C Assignments of Two New Macrocyclic Lactones Isolated from Streptomyces sp. 211726 and Revised Assignments of Azalomycins f3a, f4a and f5a. Magnetic Resonance in Chemistry. 2011. Vol. 49:30–37

            31. Yuan G, Hong K, Lin H, She Z, Li J. New Azalomycin F Analogs From Mangrove Streptomyces sp. 211726 with Activity against Microbes and Cancer Cells. Marine Drugs. 2013. Vol. 11:817–829

            32. Saha S, Zhang W, Zhang G, Zhu Y, Chen Y, Liu W, et al.. Activation and Characterization of a Cryptic Gene Cluster Reveals a Cyclization Cascade for Polycyclic Tetramate Macrolactams. Chemical Science. 2017. Vol. 8:1607–1612

            33. Moon K, Cui J, Kim E, Riandi ES, Park SH, Byun WS, et al.. Structures and Biosynthetic Pathway of Pulvomycins B–D: 22-Membered Macrolides from an Estuarine Streptomyces sp. Organic Letters. 2020. Vol. 22:5358–5362

            34. Ye X, Anjum K, Song T, Wang W, Yu S, Huang H. A New Curvularin Glycoside and its Cytotoxic and Antibacterial Analogues from Marine Actinomycete Pseudonocardia sp. HS7. Natural Product Research. 2016. Vol. 30:1156–1161

            35. Messaoudi O, Sudarman E, Bendahou M, Jansen R, Stadler M, Wink J. Kenalactams A–E, Polyene Macrolactams Isolated from Nocardiopsis CG3. Journal of Natural Products. 2019. Vol. 82:1081–1088

            36. Nie YL, Wu YD, Wang CX, Lin R, Xie Y, Fang DS. Structure Elucidation and Antitumour Activity of a New Macrolactam Produced by Marine-Derived Actinomycete Micromonospora sp. FIM05328. Natural Product Research. 2018. Vol. 32:2133–2138

            37. Fei P, Chuan-Xi W, Yang X, Hong-Lei J, Lu-Jie C, Paulina U, et al.. A New 20-Membered Macrolide Produced by a Marine-Derived Micromonospora strain. Natural Product Research. 2013. Vol. 27:1366–1371

            38. Xiangui M, Liping W, Dongyang W, Jie F, Weiming Z. Polycyclic Tetramate Macrolactams from the Marine-Derived Actinoalloteichus cyanogriseus WH1-2216-6. Chinese Journal of Organic Chemistry. 2017. Vol. 37:2352–2360

            39. Motohashi K, Takagi M, Shin-Ya K. Tetracenoquinocin and 5-iminoaranciamycin from a Sponge-derived Streptomyces sp. Sp080513GE-26. Journal of Natural Products. 2010. Vol. 73:755–758

            40. Ueda J, Khan ST, Takagi M, Shin-ya K. JBIR-58, a New Salicylamide Derivative, Isolated from a Marine Sponge-Derived Streptomyces sp. SpD081030ME-02. The Journal of Antibiotics. 2010. Vol. 63:267–269

            41. Schneemann I, Kajahn I, Ohlendorf B, Zinecker H, Erhard A, Nagel K, et al.. Mayamycin, a Cytotoxic Polyketide from a Streptomyces Strain Isolated from the Marine Sponge Halichondria panacea . Journal of Natural Products. 2010. Vol. 73:1309–1312

            42. Supong K, Thawai C, Suwanborirux K, Choowong W, Sumalee S, Pittayakhajonwut P. Antimalarial and Antitubercular C-Glycosylated benz[α]anthraquinones from the Marine-Derived Streptomyces sp. BCC45596. Phytochemistry Letters. 2012. Vol. 5:651–656

            43. Vicente J, Stewart AK, van Wagoner RM, Elliott E, Bourdelais AJ, Wright JLC. Monacyclinones, New Angucyclinone Metabolites Isolated from Streptomyces sp. M7-15 Associated with the Puerto Rican Sponge Scopalina ruetzleri . Marine Drugs. 2015. Vol. 13:4682–4700

            44. Fu P, Kong F, Wang Y, Liu P, Zuo G, Zhu W. Antibiotic Metabolites from the Coral-Associated Actinomycete Streptomyces sp. OUCMDZ-1703. Chinese Journal of Chemistry. 2013. Vol. 31:100–104

            45. Che Q, Tan H, Han X, Zhang X, Gu Q, Zhu T, et al.. Naquihexcin A, a S-Bridged Pyranonaphthoquinone Dimer Bearing an Unsaturated Hexuronic Acid Moiety from a Sponge-Derived Streptomyces sp. HDN-10-293. Organic Letters. 2016. Vol. 18:3358–3361

            46. Marchbank DH, Ptycia-Lamky VC, Decken A, Haltli BA, Kerr RG. Guanahanolide A, a Meroterpenoid with a Sesterterpene Skeleton from Coral-Derived Streptomyces sp. Organic Letters. 2020. Vol. 22:6399–6403

            47. Chen M, Chai W, Song T, Ma M, Lian XY, Zhang Z. Antiglioma Natural Products Downregulating Tumor Glycolytic Enzymes from Marine Actinomycete Streptomyces sp. ZZ406. Scientific Reports. 2018. Vol. 8:1–10

            48. Phipps RK, Blunt JW, Cole ALJ, Munro MHG. Anthracycline Derivatives from a Marine-Derived New Zealand Streptomycete . Arkivoc. 2004. Vol. 10:94–100

            49. Huang YF, Tian L, Fu HW, Hua HM, Pei YH. One New Anthraquinone from Marine Streptomyces sp. FX-58. Natural Product Research. 2006. Vol. 20:1207–1210

            50. Maskey RP, Helmke E, Fiebig HH, Laatsch H. Parimycin: Isolation and Structure Elucidation of a Novel Cytotoxic 2,3-Dihydroquinizarin Analogue of γ-Indomycinone from a Marine Streptomycete isolate. The Journal of Antibiotics. 2002. Vol. 55:1031–1035

            51. Li F, Maskey RP, Qin S, Sattler I, Fiebig HH, Maier A, et al.. Chinikomycins A and B: Isolation, Structure Elucidation, and Biological Activity of Novel Antibiotics from a Marine Streptomyces sp. Isolate M045. Journal of Natural Products2005. Vol. 68:349–353

            52. Martin GDA, Tan LT, Jensen PR, Dimayuga RE, Fairchild CR, Raventos-Suarez C, et al.. Marmycins A and B, Cytotoxic Pentacyclic C-Glycosides from a Marine Sediment-Derived Actinomycete Related to the Genus Streptomyces . Journal of Natural Products. 2007. Vol. 70:1406–1409

            53. Sato S, Iwata F, Yamada S, Kawahara H, Katayama M. Usabamycins A–C: New Anthramycin-Type Analogues from a Marine-Derived Actinomycete. Bioorganic & Medicinal Chemistry Letters. 2011. Vol. 21:7099–7101

            54. Zhang H, Wang H, Cui H, Li Z, Xie Z, Pu Y, et al.. A New Anthracene Derivative from Marine Streptomyces sp. W007 Exhibiting Highly and Selectively Cytotoxic Activities. Marine Drugs. 2011. Vol. 9:1502–1509

            55. Huang H, Yang T, Ren X, Liu J, Song Y, Sun A, et al.. Cytotoxic Angucycline Class Glycosides from the Deep Sea Actinomycete Streptomyces lusitanus SCSIO LR32. Journal of Natural Products. 2012. Vol. 75:202–208

            56. Fu P, Johnson M, Chen H, Posner BA, MacMillan JB. Carpatamides A–C, Cytotoxic Arylamine Derivatives from a Marine-Derived Streptomyces sp. Journal of Natural Products. 2014. Vol. 77:1245–1248

            57. Fu S, Wang F, Li H, Bao Y, Yang Y, Shen H. Secondary Metabolites from Marine-Derived Streptomyces Antibioticus Strain H74-21. Natural Product Research. 2016. Vol. 30:2460–2467

            58. Song Y, Liu G, Li J, Huang H, Zhang X, Zhang H, et al.. Cytotoxic and Antibacterial Angucycline-and Prodigiosin-Analogues from the Deep-Sea Derived Streptomyces sp. SCSIO 11594. Marine Drugs. 2015. Vol. 13:1304–1316

            59. Tsukahara K, Toume K, Ishikawa N, Abdelfattah MS, Ishibashi M. Novel Cytotoxic Isobenzofuran Derivatives from Streptomyces sp. IFM 11490. Tetrahedron Letters. 2015. Vol. 56:6345–6347

            60. Liang Y, Xie X, Chen L, Yan S, Ye X, Anjum K, et al.. Bioactive Polycyclic Quinones from Marine Streptomyces sp. 182SMLY. Marine Drugs. 2016. Vol. 14:10

            61. Mullowney MW, Ó hAinmhire E, Shaikh A, Wei X, Tanouye U, Santarsiero BD, et al.. Diazaquinomycins E-G, Novel Diaza-Anthracene Analogs from a Marine-Derived Streptomyces sp. Marine Drugs. 2014. Vol. 12:3574–3586

            62. Xie Z, Zhou L, Guo L, Yang X, Qu G, Wu C, et al.. Grisemycin, a Bridged Angucyclinone with a Methylsulfinyl Moiety from a Marine-Derived Streptomyces sp. Organic Letters. 2016. Vol. 18:1402–1405

            63. He H, Ding WD, Bernan VS, Richardson AD, Ireland CM, Greenstein M, et al.. Lomaiviticins A and B, Potent Antitumor Antibiotics from Micromonospora lomaivitiensis . Journal of the American Chemical Society. 2001. Vol. 123:5362–5363

            64. Wang Y, Wang L, Zhuang Y, Kong F, Zhang C, Zhu W. Phenolic Polyketides from the Co-Cultivation of Marine-Derived Penicillium sp. WC-29-5 and Streptomyces fradiae 007. Marine Drugs. 2014. Vol. 12:2079–2088

            65. Zhu X, Duan Y, Cui Z, Wang Z, Li Z, Zhang Y, et al.. Cytotoxic Rearranged Angucycline Glycosides from Deep Sea-Derived Streptomyces lusitanus SCSIO LR32. The Journal of Antibiotics (Tokyo). 2017. Vol. 70:819–822

            66. Lai Z, Yu J, Ling H, Song Y, Yuan J, Ju J, et al.. Grincamycins I-K, Cytotoxic Angucycline Glycosides Derived from Marine-Derived Actinomycete Streptomyces lusitanus SCSIO LR32. Planta Medica. 2018. Vol. 84:201–207

            67. Jiang YJ, Zhang DS, Zhang HJ, Li JQ, Ding WJ, Xu CD, et al.. Medermycin-Type Naphthoquinones from the Marine-Derived Streptomyces sp. XMA39. Journal of Natural Products. 2018. Vol. 81:2120–2124

            68. Mullowney MW, ÓhAinmhire E, Tanouye U, Burdette JE, Pham VC, Murphy BT. A Pimarane Diterpene and Cytotoxic Angucyclines from a Marine-Derived Micromonospora sp. in Vietnam’s East Sea. Marine Drugs. 2015. Vol. 13:5815–5827

            69. Fang Z, Jiang X, Zhang Q, Zhang L, Zhang W, Yang C, et al.. S-Bridged Thioether and Structure-Diversified Angucyclinone Derivatives from the South China Sea-Derived Micromonospora echinospora SCSIO 04089. Journal of Natural Products. 2020. Vol. 83:3122–3130

            70. Gan M, Liu B, Tan Y, Wang Q, Zhou H, He H, et al.. Saccharothrixones A-D, Tetracenomycin-Type Polyketides from the Marine-Derived Actinomycete Saccharothrix sp. 10-10. Journal of Natural Products. 2015. Vol. 78:2260–2265

            71. Yang T, Yamada K, Zhou T, Harunari E, Igarashi Y, Yerahara T, et al.. Akazamicin, a Cytotoxic Aromatic Polyketidefrom Marine-Derived Nonomuraea sp. The Journal of Antibiotics. 2019. Vol. 72:202–209

            72. Williams DE, Dalisay DS, Li F, Amphlett J, Maneerat W, Ghavez MAG, et al.. Nahuoic Acid A Produced by a Streptomyces sp. Isolated from a Marine Sediment is a Selective SAM-Competitive Inhibitor of the Histone Methyltransferase SETD8. Organic Letters. 2013. Vol. 15:414–417

            73. Williams DE, Izard F, Arnould S, Dalisay DS, Tantapakul C, Maneerat W, et al.. Structures of Nahuoic Acids B–E Produced in Culture by a Streptomyces sp. Isolated from a Marine Sediment and Evidence for the Inhibition of the Histone Methyl Transferase SETD8 in Human Cancer Cells by Nahuoic Acid A. The Journal of Organic Chemistry. 2016. Vol. 81:1324–1332

            74. Xie XC, Mei WL, Zhao YX, Hong K. A New Degraded Sesquiterpene from Marine Actinomycete Streptomyces sp. 0616208. Chinese Chemical Letters. 2006. Vol. 17:1463–1465

            75. Hayakawa Y, Shirasaki S, Shiba S, Kawasaki T, Matsuo Y, Adachi K, et al.. Piericidins C7 and C8, New Cytotoxic Antibiotics Produced by a Marine Streptomyces sp. The Journal of Antibiotics. 2007. Vol. 60:196–200

            76. Manam RR, Teisan S, White DJ, Nicholson B, Grodberg J, Neuteboom STC, et al.. Lajollamycin, a Nitro-Tetraene Spiro-β-Lactone-γ-Lactam Antibiotic from the Marine Actinomycete Streptomyces nodosus. Journal of Natural Products. 2005. Vol. 68:240–243

            77. Han X, Liu Z, Zhang Z, Zhang X, Zhu T, Gu Q, et al.. Geranylpyrrol A and Piericidin F from Streptomyces sp. CHQ-64 ArdmF. Journal of Natural Products. 2017. Vol. 80:1684–1687

            78. Schleissner C, Pérez M, Losada A, Rodríguez P, Crespo C, Zúñiga P, et al.. Antitumor Actinopyranones Produced by Streptomyces albus POR-04-15-053 Isolated from a Marine Sediment. Journal of Natural Products. 2011. Vol. 74:1590–1596

            79. Shaaban KA, Helmke E, Kelter G, Fiebig HH, Laatsch H. Glucopiericidin C: A Cytotoxic Piericidin Glucoside Antibiotic Produced by a Marine Streptomyces isolate. The Journal of Antibiotics. 2011. Vol. 64:205–209

            80. Igarashi Y, Asano D, Furihata K, Oku N, Miyanaga S, Sakurai H, et al.. Absolute Configuration of Pterocidin, a Potent Inhibitor of Tumor Cell Invasion from a Marine-Derived Streptomyces . Tetrahedron Letters. 2012. Vol. 53:654–656

            81. Bae M, Park SH, Kwon Y, Lee SK, Shin J, Nam JW, et al.. QM-HiFSA-aided Structure Determination of Succinilenes AD, New Triene Polyols from a Marine-Derived Streptomyces sp. Marine drugs. 2017. Vol. 15:E38

            82. Huang H, Cao Y, Tian L, Lin W, Zhang K. A New Polyunsaturated Acid from the Marine-Derived Streptomyces violans (No. HTTA-F04129). Chemistry of Natural Compounds. 2014. Vol. 50:402–404

            83. Bae M, Kim H, Shin Y, Kim BY, Lee SK, Oh KB, et al.. Separacenes A–D, Novel Polyene Polyols from the Marine Actinomycete, Streptomyces sp. Marine Drugs. 2013. Vol. 11:2882–2893

            84. Asolkar RN, Jensen PR, Kauffman CA, Fenical W. Daryamides A−C, Weakly Cytotoxic Polyketides from a Marine-Derived Actinomycete of the Genus Streptomyces Strain CNQ-085. Journal of Natural Products. 2006. Vol. 69:1756–1759

            85. Sato S, Iwata F, Mukai T, Yamada S, Takeo J, Abe A, et al.. Indoxamycins A−F. Cytotoxic Tricycklic Polypropionates from a Marine-Derived Actinomycete. The Journal of Organic Chemistry. 2009. Vol. 74:5502–5509

            86. Jiang YJ, Li JQ, Zhang HJ, Ding WJ, Ma ZJ. Cyclizidine Type Alkaloids from Streptomyces sp. HNA39. Journal of Natural Products. 2018. Vol. 81:394–399

            87. Kawahara T, Itoh M, Izumikawa M, Kozone I, Sakata N, Tsuchida T, et al.. New Hydroxamate Metabolite, MBJ-0003 from Micromonospora sp. 29867. Journal of Antibiotics. 2014. Vol. 67:261–263

            88. Sarmiento-Vizcalno A, Braña AF, Pérez-Victoria I, Martín J, Pedro ND, Cruz MD, et al.. Paulomycin G, a New Naturalproduct with Cytotoxic Activity Against Tumor Cell Lines Produced by Deep-Sea Sediment Derived Micromonospora matsumotoense M-412 from the Aviles Canyon in the Cantabrian Sea. Marine Drugs. 2017. Vol. 15:271–279

            89. Zhang S, Xie Q, Sun C, Tian XP, Gui C, Qin X, et al.. Cytotoxic Kendomycins Containing the Carbacylicansa Scaffold from the Marine-Derived Verrucosispora sp. SCSIO 07399. Journal of Natural Products. 2019. Vol. 82:3366–3371

            90. Zhen X, Gong T, Liu F, Zhang PC, Zhou WQ, Li Y, et al.. A New Analogue of Echinomycin and a New Cyclic Dipeptide from a Marine-Derived Streptomyces sp. LS298. Marine Drugs. 2015. Vol. 13:6947–6961

            91. Cheng C, Othman EM, Stopper H, Edrada-Ebel R, Hentschel U, Abdelmohsen UR. Isolation of Petrocidin A, a New Cytotoxic Cyclic Dipeptide from the Marine Sponge-Derived Bacterium Streptomyces sp. SBT348. Marine Drugs. 2017. Vol. 15:E383

            92. Um S, Kim YJ, Kwon H, Wen H, Kim SH, Kwon HC, et al.. Sungsanpin, a Lasso Peptide from a Deep-Sea Streptomycete . Journal of Natural Products. 2013. Vol. 76:873–879

            93. Um S, Choi TJ, Kim H, Kim BY, Kim SH, Lee SK, et al.. Ohmyungsamycins A and B: Cytotoxic and Antimicrobial Cyclic Peptides Produced by Streptomyces sp. from a Volcanic Island. The Journal of Organic Chemistry. 2013. Vol. 78:12321–12329

            94. Ye X, Anjum K, Song T, Wang W, Liang Y, Chen M, et al.. Antiproliferative Cyclodepsipeptides from the Marine Actinomycete Streptomyces sp. P11-23B Downregulating the Tumor Metabolic Enzymes of Glycolysis, Glutaminolysis, and Lipogenesis. Phytochemistry. 2017. Vol. 135:151–159

            95. Aftab U, Sajid I. Antitumor Peptides from Streptomyces sp. SSA 13, Isolated from Arabian Sea. International Journal of Peptide Research and Therapeutics. 2017. Vol. 23:199–211

            96. Ou YX, Huang JF, Li XM, Kang QJ, Pan YT. Three New 2,5-Diketopiperazines from the Fish Intestinal Streptomyces sp. MNU FJ-36. Natural Product Research. 2016. Vol. 30:1771–1775

            97. Shaala LA, Youssef DT, Badr JM, Harakeh SM. Bioactive 2(1H)-Pyrazinones and Diketopiperazine Alkaloids from a Tunicatederived Actinomycete Streptomyces sp. Molecules. 2016. Vol. 21:E1116

            98. Ibrahim AH, Attia EZ, Hajjar D, Anany NA, Desoukey SY, Fouad MA, et al.. New Cytotoxic Cyclic Peptide from the Marine Sponge-Associated Nocardiopsis sp. UR67. Marine Drugs. 2018. Vol. 16:290

            99. Cho JY, Williams PG, Kwon HC, Jensen PR, Fenical W. Lucentamycins A−D, Cytotoxic Peptides from the Marine-Derived Actinomycete Nocardiopsislucentensis . Journal of Natural Products. 2007. Vol. 70:1321–1328

            100. Sun M, Chen X, Li C, Lu C, Shen Y. New Diketopiperazine Derivatives with Cytotoxicity from Nocardiopsis sp. YIM M13066. Journal of Antibiotics. 2017. Vol. 70:795–797

            101. Liu D, Lin H, Proksch P, Tang X, Shao Z, Lin W. Microbacterins A and B, New Peptaibols from the Deep Sea Actinomycete Microbacterium sediminis sp. Nov. YLB-01(T). Organic Letter. 2015. Vol. 17:1220–1223

            102. Miller ED, Kauffman CA, Jensen PR, Fenical W. Piperazimycins: Cytotoxic Hexadepsipeptides from a Marine-Derived Bacterium of the Genus Streptomyces . The Journal of Organic Chemistry. 2007. Vol. 72:323–330

            103. Igarashi Y, Shimasaki R, Miyanaga S, Oku N, Onaka H, Sakurai H, et al.. Rakicidin D, an Inhibitor of Tumor Cell Invasion from Marine-Derived Streptomyces sp. The Journal of Antibiotics. 2010. Vol. 63:563–565

            104. Williams DE, Dalisay DS, Patrick BO, Matainaho T, Andrusiak K, Deshpande R, et al.. Padanamides A and B, Highly Modified Linear Tetrapeptides Produced in Culture by a Streptomyces sp. Isolated from a Marine Sediment. Organic Letters. 2011. Vol. 13:3936–3939

            105. Nair V, Kim MC, Golen JA, Rheingold AL, Castro GA, Jensen PR, et al.. Verrucosamide A Cytotoxic 1, 4-Thiazepane-Containing Thiodepsipeptide from a Marine-Derived Actinomycete. Marine Drugs. 2020. Vol. 18:549

            106. Ding L, Pfoh R, Ruhl S, Qin S, Laatsch H. T-Muurolol Sesquiterpenes from the Marine Streptomyces sp. M491 and Revision of the Configuration of Previously Reported Amorphanes. Journal of Natural Products. 2009. Vol. 72:99–101

            107. Zhang YM, Li HY, Hu C, Sheng HF, Zhang Y, Lin B, et al.. Ergosterols from the Culture Broth of Marine Streptomyces anandii H41-59. Marine Drugs. 2016. Vol. 14:84

            108. Simmons L, Kaufmann K, Garcia R, Schwar G, Huch V, Muller R. Bendigoles D-F, Bioactive Sterols from the Marine Sponge-Derived Actinomadura sp. SBMs009. Bioorganic & Medicinal Chemistry. 2011. Vol. 19:6570–6575

            109. Izumikawa M, Khan ST, Komaki H, Takagi M, Shin-ya K. JBIR-31, a New Teleocidin Analog, Produced by Salt-Requiring Streptomyces sp. NBRC 105896 Isolated from a Marine Sponge. The Journal of Antibiotics. 2010. Vol. 63:33–36

            110. Che Q, Li J, Li D, Gu Q, Zhu T. Structure and Absolute Configuration of DrimentineI, An Alkaloid from Streptomyces sp. CHQ-64[J]. The Journal of Antibiotics. 2016. Vol. 69:467–469

            111. Nam SJ, Kauffman CA, Paul LA, Jensen PR, Fenical W. Actinoranone, A Cytotoxic Meroterpenoid of Unprecedented Structure from a Marine Adapted Streptomyces sp. Organic Letters. 2013. Vol. 15:5400–5403

            112. Kwon HC, Espindola APDM, Park JS, Prieto-Davo AP, Rose M, Jensen PR, et al.. Nitropyrrolins A-E, Cytotoxic Farnesyl-α-Nitropyrroles from a Marine-Derived Bacterium within the Actinomycete Family Streptomycetaceae. Journal of Natural Products. 2010. Vol. 73:2047–2052

            113. Song Y, Huang H, Chen Y, Ding J, Zhang Y, Sun A, et al.. Cytotoxic and Antibacterial Marfuraquinocins from the Deep South China Sea-Derived Streptomyces niveus SCSIO 3406. Journal of Natural Products. 2013. Vol. 76:2263–2268

            114. Cheng YB, Jensen PR, Fenical W. Cytotoxic and Antimicrobial Napyradiomycins from Two Marine-Derived Streptomyces Strains. European Journal of Organic Chemistry. 2013. Vol. 2013:3751–3757

            115. Farnaes L, Coufal NG, Kauffman CA, Rheingold AL, Dipasquale AG, Jensen PR, et al.. Napyradiomycin Derivatives, Produced by a Marine-Derived Actinomycete, Illustrate Cytotoxicity by Induction of Apoptosis. Journal of Natural Products. 2014. Vol. 77:15–21

            116. Macherla VR, Liu J, Bellows C, Teisan S, Nicholson B, Lam KS, et al.. Glaciapyrroles A, B and C, Pyrrolosesquiterpenes from a Streptomyces sp. Isolated from an Alaskan Marine Sediment. Journal of Natural Products. 2005. Vol. 68:780–783

            117. Sánchez López JM, Martínez Insua M, Pérez Baz J, Fernandez Puentes JL, Canedo Hernandez LM. New Cytotoxic Indolic Metabolites from a Marine Streptomyces . Journal of Natural Products. 2003. Vol. 66:863–864

            118. Cheng C, Othman EM, Fekete A, Krischke M, Stopper H, Edrada-Ebel R, et al.. Strepoxazine A, a New Cytotoxic Phenoxazin from the Marine Sponge-Derived Bacterium Streptomyces sp. SBT345. Tetrahedron Letters. 2016. Vol. 57:4196–4199

            119. Ding L, Münch J, Goerls H, Maier A, Fiebig HH, Lin WH, et al.. Xiamycin a Pentacyclic Indolosesquiterpene with Selective Anti-HIV Activity from a Bacterial Mangrove Endophyte. Bioorganic & Medicinal Chemistry Letters. 2010. Vol. 20:6685–6687

            120. Takahashi A, Kurasawa S, Ikeda D, Okami Y, Takeuchi T. Altemicidin, a New Acaricidal and Antitumor Substance I. Taxonomy, Fermentation, Isolation and Physico-Chemical and Biological Properties. The Journal of Antibiotics. 1989. Vol. 42:1556–1561

            121. Chen L, Chai W, Wang W, Song T, Lian XY, Zhang Z. Cytotoxic Bagremycins from Mangrove-Derived Streptomyces sp. Q22. Journal of Natural Products. 2017. Vol. 80:1450–1456

            122. Jeong SY, Shin HJ, Kim TS, Lee HS, Park S, Kim HM. Streptokordin, a New Cytotoxic Compound of the Methylpyridine Class from a Marine-Derived Streptomyces sp. KORDI-3238. The Journal of Antibiotics. 2006. Vol. 59:234–240

            123. Hughes CC, Prieto-Davo A, Jensen PR, Fenical W. The Marinopyrroles, Antibiotics of an Unprecedented Structure Class from a Marine Streptomyces sp. Organic Letters. 2008. Vol. 10:629–631

            124. Hughes CC, Kauffman CA, Jensen PR, Fenical W. Structures, Reactivities and Antibiotic Properties of the Marinopyrroles A−F. The Journal of Organic Chemistry. 2010. Vol. 75:3240–3250

            125. Boonlarppradab C, Kauffman CA, Jensen PR, Fenical W. Marineosins A and B, Cytotoxic Spiroaminals from a Marine-D Actinomycete. Organic Letters. 2008. Vol. 10:5505–5508

            126. Hughes CC, MacMillan JB, Gaudêncio SP, Jensen PR, Fenical W. The Ammosamides: Structures of Cell Cycle Modulators from a Marine-Derived Streptomyces Species. Angewandte Chemie International Edition. 2009. Vol. 48:725–727

            127. Hawas UW, Shaaban M, Shaaban KA, Speitling M, Maier A, Kelter G, et al.. Mansouramycins A−D, Cytotoxic Isoquinolinequinones from a Marine Streptomycete . Journal of Natural Products. 2009. Vol. 72:2120–2124

            128. Li B, Chen G, Bai J, Jing YK, Pei YH. A Bisamide and Four Diketopiperazines from a Marine-Derived Streptomyces sp. Journal of Asian Natural Products Research. 2011. Vol. 13:1146–1150

            129. Fu P, Zhuang Y, Wang Y, Liu P, Qi X, Gu K, et al.. New Indolocarbazoles from a Mutant Strain of the Marine-Derived Actinomycete 007M135. Organic Letters. 2012. Vol. 14:6194–6197

            130. Zhang W, Liu Z, Li S, Yang T, Zhang Q, Ma L, et al.. Spiroindimicins A–D: New Bisindole Alkaloids from a Deep-Sea-Derived Actinomycete. Organic Letters. 2012. Vol. 14:3364–3367

            131. Wu SJ, Fotso S, Li F, Qin S, Kelter G, Fiebig HH, et al.. N-Carboxamido-Staurosporine and Selina-4 (14), 7 (11)-Diene-8, 9-Diol, New Metabolites from a Marine Streptomyces sp. The Journal of Antibiotics. 2006. Vol. 59:331–337

            132. Zhang W, Ma L, Li S, Liu Z, Chen Y, Zhang H, et al.. Indimicins A–E, Disindole Alkaloids from the Deep-Sea-Derived Streptomyces sp. SCSIO 03032. Journal of Natural Products2014. Vol. 77:1887–1892

            133. Pan E, Jamison M, Yousufuddin M, MacMillan JB. Ammosamide D, an Oxidatively Ring Opened Ammosamide Analog from a Marine-Derived Streptomyces variabilis . Organic Letters. 2012. Vol. 14:2390–2393

            134. Fu P, Yang C, Wang Y, Liu P, Ma Y, Xu L, et al.. Streptocarbazoles A and B, Two Novel Indolocarbazoles from the Marine-Derived Actinomycete Strain Streptomyces sp. FMA. Organic Letters. 2012. Vol. 14:2422–2425

            135. Abdelfattah MS. A New Dioactive Aminophenoxazinone Alkaloid from a Marine-Derived Actinomycete. Natural Product Research. 2013. Vol. 27:2126–2131

            136. Kondratyuk TP, Park EJ, Yu R, Van Breemen RB, Asolkar RN, Murphy BT, et al.. Novel Marine Phenazines as Potential Cancer Chemopreventive and Anti-Inflammatory Agents. Marine Drug. 2012. Vol. 10:451–464

            137. Fu P, Legako A, La S, MacMillan JB. Discovery, Characterization, and Analogue Synthesis of Bohemamine Dimers Generated by Non-Enzymatic Biosynthesis. Chemistry–A European Journal. 2016. Vol. 22:3491–3495

            138. Alvarez-Mico X, Jensen PR, Fenical W, Hughes CC. Chlorizidine, A Cytotoxic 5H-Pyrrolo[2,1-a]isoindol-5-one-Containing Alkaloid from a Marine Streptomyces sp. Organic Letters. 2013. Vol. 15:988–991

            139. Fu P, Zhu Y, Mei X, Wang Y, Jia H, Zhang C, et al.. Acyclic Congeners from Actinoalloteichus Cyanogriseus Provide Insights into Cyclic Bipyridine Glycoside Formation. Organic Letters. 2014. Vol. 16:4264–4267

            140. El-Hawary SS, Sayed AM, Mohammed R, Khanfar MA, Rateb ME, Mohammed TA, et al.. New Pim-1 Kinase Inhibitor from the Co-Culture of Two Sponge-Associated Actinomycetes. Frontiers in Chemistry. 2018. Vol. 6:538

            141. Silva AET, Guimaraes LA, Ferreira EG, Torres MCM, Silva AB, Branco PC, et al.. Bioprospecting Anticancer Compounds From the Marine-Derived Actinobacteria Actinomadura sp. Collected at the Saint Peter and Saint Paul Archipelago (Brazil). Journal of the Brazilian Chemical Society. 2017. Vol. 28:465–474

            142. Kwon Y, Kim SH, Shin Y, Bae M, Kim BY, Lee SK, et al.. A New Benzofuran Glycoside and Indole Alkaloids from a Sponge-Associated Rare Actinomycete, Amycolatopsis sp. Marine Drugs. 2014. Vol. 12:2326–2340

            143. Qin L, Yi W, Lian XY, Zhang Z. Bioactive Alkaloids from the Actinomycete Actinoalloteichus sp. ZZ1866. Journal of Natural Products. 2020. Vol. 83:2686–2695

            144. Subramani R, Aalbersberg W. Marine Actinomycetes: An Ongoing Source of Novel Bioactive Metabolites. Microbiological Research. 2012. Vol. 167:571–580

            145. Olano C, Méndez C, Salas JA. Antitumor Compounds from Marine Actinomycetes. Marine Drugs. 2009. Vol. 7:210–248

            146. Wang C, Du W, Lu H, Lan J, Liang K, Cao S. A Review: Halogenated Compounds from Marine Actinomycetes. Molecules. 2021. Vol. 26:2754

            147. Wang C, Lu Y, Cao S. Antimicrobial Compounds from Marine Actinomycetes. Archives of Pharmacal Research. 2020. Vol. 43:677–704

            148. Uz Zaman KHA, Park JH, DeVine L, Hu Z, Wu X, Kim HS, et al.. Secondary Metabolites from the Leather Coral-Derived Fungal Strain Xylaria sp. FM1005 and Their Glycoprotein IIb/IIIa Inhibitory Activity. Journal of Natural Products. 2021. Vol. 84:466–473

            149. Colis LC, Woo CM, Hegan DC, Li Z, Glazer PM, Herzon SB. The Cytotoxicity of (-)-Lomaiviticin A Arises from Induction of Double-Strand Breaks in DNA. Nature Chemistry. 2014. Vol. 6:504–510

            150. Hirata Y, Uemura D. Halichondrins - antitumor polyether macrolides from a marine sponge. Pure and Applied Chemistry. 1986. Vol. 58:701–710

            151. Yu MJ, Zheng W, Seletsk BM. From micrograms to grams: scale-up synthesis of eribulin mesylate. Natural Product Reports. 2013. Vol. 30:1158–1164

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            15 November 2022
            : 1
            : 4
            : 445-475
            Affiliations
            [a ]Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China
            [b ]Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, 200 W. Kawili St., Hilo, HI 96720, USA
            Author notes
            *Correspondence: wangcong123206@ 123456163.com (C. Wang); scao@ 123456hawaii.edu (S. Cao)

            1Ziyan Qiu and Yinshuang Wu contributed to the manuscript equally.

            Article
            10.15212/AMM-2022-0028
            36588746
            66879633-43ef-451c-b41c-9ff513d25286
            Copyright © 2022 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 11 August 2022
            : 16 October 2022
            : 28 October 2022
            Page count
            Figures: 10, Tables: 1, References: 151, Pages: 31
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            marine actinomycetes,antitumor bioactivity,chemical structures,marine natural products

            Comments

            Comment on this article