Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2412 2023-08-09 20:52:40 |
2 format change Meta information modification 2412 2023-08-10 03:07:48 | |
3 format change -1 word(s) 2411 2023-08-10 03:08:27 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Trinidad-Calderón, P.A.; Varela-Chinchilla, C.D.; García-Lara, S. Depsipeptides Targeting Tumor Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/47855 (accessed on 27 July 2024).
Trinidad-Calderón PA, Varela-Chinchilla CD, García-Lara S. Depsipeptides Targeting Tumor Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/47855. Accessed July 27, 2024.
Trinidad-Calderón, Plinio A., Carlos Daniel Varela-Chinchilla, Silverio García-Lara. "Depsipeptides Targeting Tumor Cells" Encyclopedia, https://encyclopedia.pub/entry/47855 (accessed July 27, 2024).
Trinidad-Calderón, P.A., Varela-Chinchilla, C.D., & García-Lara, S. (2023, August 09). Depsipeptides Targeting Tumor Cells. In Encyclopedia. https://encyclopedia.pub/entry/47855
Trinidad-Calderón, Plinio A., et al. "Depsipeptides Targeting Tumor Cells." Encyclopedia. Web. 09 August, 2023.
Depsipeptides Targeting Tumor Cells
Edit

Cancer is considered one of the most threatening diseases worldwide. Diet could be one of the factors that can be enhanced to comprehensively address a cancer patient’s condition. Unfortunately, most molecules capable of targeting cancer cells are found in uncommon food sources. Among them, depsipeptides have emerged as one of the most reliable choices for cancer treatment. These cyclic amino acid oligomers, with one or more subunits replaced by a hydroxylated carboxylic acid resulting in one lactone bond in a core ring, have broadly proven their cancer-targeting efficacy, some even reaching clinical trials and being commercialized as “anticancer” drugs.

depsipeptide anticancer cancer-targeting cell functional food enrichment

1. Introduction

Nowadays, cancer is considered the first or second major cause of death in multiple countries around the world [1][2]. In 2020, more than 19 million new cases and nearly 10 million deaths associated with this disease were reported [3][4][5]. Unsurprisingly, cancer cases are expected to reach the 24 million mark by 2035, and their incidence to triple by 2050 [6][7].
Although recognized cancer therapies are being further enhanced [8], the consumption of enriched foods to boost the recovery of cancer patients is also gaining attention [9]. To date, nearly 35% of chemotherapeutic drugs are nature-derived products, making these an important source of oncological pharmaceuticals [10]. Nonetheless, only a limited number of recognized molecules found in foods, especially in animals, have been shown to possess promising cancer cell-targeting abilities in preclinical trials [11].
Among the spectrum of potential therapeutic molecules, the tumor-targeting capability of natural cyclic peptides has been deeply explored [12][13]. Scientifically known as depsipeptides, these peptide cyclo-oligomers are characterized by having one or more amino acids replaced by a hydroxylated carboxylic acid, resulting in one lactone bond in a core ring (Figure 1) [14]. Interestingly, they account for more than 1300 natural molecules reported to contain different acyl groups and other moieties [15][16] as well as to perform varied bioactivities [17][18].
Figure 1. Peptide bond structure of depsipeptides. Lactone bond in the core ring marked in red.
Hitherto, few authors have recognized that the vast majority of depsipeptides are found in marine species, most of which are not present in easily accessible areas and are hard to reproduce in the laboratory environment without proper instruction [10][19]. Some other authors have highlighted that the knowledge on “anticancer” mechanisms, known limitations, and faced challenges of using these molecules is minimal [20].

2. Depsipeptides with a Recognized Mechanism of Targeting Cancer Cells

Depsipeptides induce cytotoxicity by various mechanisms, with their principal features being the capacity to perforate cell membranes and tubulin–microtubule imbalance, eventually inducing apoptosis [21].

2.1. Depsipeptides Inducing Apoptosis in Tumor Cells

Apoptosis, as the most prevalent and well-defined form of programmed cell death [22], plays an important role in regulating tumor cells [23]. Unsurprisingly, resistance to apoptosis—due to the inactivation or loss of caspases [24] and dysregulation of the mitochondrial pathway [25]—is considered a major advantage for oncogenic cells [26].
In general, these depsipeptides potently activate caspase-3, Fas/FasL, p21, and p27, and inhibit histone deacetylases, all of which result in pro-apoptotic signaling [27][28][29][30] Here, the researchers present the state-of-the-art on depsipeptides that induce apoptosis in tumor cells (Table 1 and Figure 2).
Figure 2. Models of depsipeptides inducing apoptosis in tumor cells. (a) Apratoxin A; (b) Aurilides; (c) Beauvericins; (d) Dehydrodidemnin B; (e) Enniatins; (f) Lagunamides; (g) Coibamide A; (h) Grassypeptolides; (i) Hantupeptins; (j) Tiahuramides.
Table 1. Formula and molecular weight of depsipeptides inducing apoptosis in tumor cells.

Key

Depsipeptide

IUPAC Condensed Formula

Molecular Weight

(g/mol)

Ref.

a

Apratoxin

Cyclo[N(Me)Ala-N(Me)Ile-Pro-Unk-Tyr(Me)]

840.1

[31]

b

Aurilide

Cyclo[N(Me)Ala-Unk-Val-D-N(Me)Leu-Sar-Val]

834.1

[21]

c

Beauvericin

Cyclo[D-Oval-N(Me)Phe-D-Oval-N(Me)Phe-D-Oval-N(Me)Phe]

783.9

[32]

d

Coibamide

N(Me2)Val-Oval-N(Me)Ser(Me)-N(Me)Leu-N(Me)Thr(1)-N(Me)Ser(Me)-N(Me)Ile-Ala-N(Me)Leu-Tyr(Me)-N(Me)Ala-(1)

1287.6

[33]

e

Dehydrodidemnin

Pyruvoyl-Pro-D-N(Me)Leu-D-Thr(1)-Unk-Leu-Pro-DL-N(Me)Tyr(Me)-(1)

1110.3

[34]

f

Enniatin

Cyclo[DL-Oval-DL-N(Me)xiIle-DL-Oval-DL-N(Me)xiIle-DL-Oval-DL-N(Me)xiIle]

681.9

[35]

g

Grassypeptolide

Cyclo[D-N(Me)Leu-D-aThr-Unk-N(Me)Val-Pro-Unk]

1102.4

[36]

h

Hantupeptin

Cyclo[N(Me)Ile-Ophe-Pro-N(Me)Val-Unk-Val]

740.9

[37]

i

Lagunamide

Cyclo[Ala-D-N(Me)Phe-Sar-aIle-N(Me)Ala-Unk]

842.1

[38]

j

Tiahuramide

Cyclo[N(Me)Ile-Unk-Val-N(Me)Val-Ophe-Pro]

736.9

[39]

2.2. Depsipeptides Inducing Autophagy in Tumor Cells

Autophagy is the process that uses autophagosomes to deliver cytoplasmic material to the lysosome for degradation [40]. Autophagy-inducing depsipeptides can activate intracellular messengers such as adenosine monophosphate-activated protein kinase and transcription factor EB [41] (Figure 3). They can also inhibit survival-inducing molecules such as hypoxia-inducible factor 1-alpha and mammalian target of rapamycin [29], thereby inducing autophagy through various mechanisms [42][43].
Figure 3. Staging model of depsipeptides inducing autophagy in tumor cells.

Beauvenniatins

Initially isolated from a mycelial extract of Acremonium spp., with additional analogs also produced by Fusarium and Isaria spp. [44], the structure of these peptides is a hybrid between the aromatic and aliphatic cyclodepsipeptides, with moieties of N-Me-valine, N-Me-leucine, and N-Me-phenylalanine (Table 2) [45].
Table 2. Formula and molecular weight of beauvenniatin, a depsipeptide triggering autophagy in tumor cells.
Beauvenniatin F has exhibited strong cytotoxicity against adriamycin-resistant myelogenous leukemia cells (K-562), with an IC50 of 2.78 μM, and autophagy-inducing activity at a concentration of 20 μM in green fluorescent protein-light chain 3-stable HeLa cells [46]. These depsipeptides also displayed cytotoxicity against epithelial carcinoma (KB), lymphoma (BC), and lung carcinoma cells (NCI-H187), with IC50 values ranging from 1.00 to 2.29 μM as well as against kidney epithelial cells (Vero) at 1.9–5.5 μM [47].

2.3. Depsipeptides Inhibiting Elastases

Elastases are a group of proteases that degrade and lyse elastin, one of the main components of the extracellular matrix [48]. These enzymes are also involved in tumor cell migration [49]. Consequently, any dysregulation of elastase may result in the genesis of various diseases such as cancer [50] as well as metastasis [49].
Here, the researchers elaborate on the depsipeptides with elastase-inhibiting potential (Table 3 and Figure 4).
Figure 4. Micropeptins (a) and tutuilamides (b), depsipeptides capable of inhibiting elastases.
Table 3. Formula and molecular weight of depsipeptides inhibiting elastases in tumor cells.

Key

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

a

Micropeptins

(3S)-4-[[(2S,5S,8S,11R,12S,15S,18S,21R)-15-(4-aminobutyl)-2-[(2S)-butan-2-yl]-21-hydroxy-5-[(4-hydroxyphenyl)methyl]-4,11-dimethyl-3,6,9,13,16,22-hexaoxo-8-propan-2-yl-10-oxa-1,4,7,14,17-pentazabiCyclo [16.3.1]docosan-12-yl]amino]-3-(hexanoylamino)-4-oxobutanoic acid

945.1

[51]

b

Tutuilamides

(2S)-N-[(2S,5S,8S,11R,12S,15Z,18S,21R)-2-benzyl-15-ethylidene-21-hydroxy-5-[(4-hydroxyphenyl)methyl]-4,11-dimethyl-3,6,9,13,16,22-hexaoxo-8-propan-2-yl-10-oxa-1,4,7,14,17-pentazabiCyclo [16.3.1]docosan-12-yl]-2-[[(2S)-2-[[I-4-chloro-3-methylbut-3-enoyl]amino]propanoyl]amino]-3-methylbutanamide

1007.6

[52]

2.4. Depsipeptides Inhibiting Histone Deacetylases

Histone deacetylases constitute a group of enzymes catalyzing the deacetylation of lysine residues found in histones and other proteins [53]. The acetylation of these residues results in a less condensed chromatin structure due to increased space between the nucleosome and DNA, thus modulating cell cycle progression [54]. Interestingly, histone deacetylase inhibitors can induce apoptosis, delay cell cycle progression, and inhibit cancer cell differentiation [30].
Here, the researchers describe depsipeptides that inhibit histone deacetylases in cancer cells (Figure 5 and Table 4).
Figure 5. Depsipeptides inhibiting histone acetyltransferases in tumor cells. (a) Bassianolide; (b) Clavatustides; (c) Cryptophycins; (d) Largazole; (e) Lyngbyabellins; (f) Romidepsin; and (g) Sansalvamide.
Table 4. Formula and molecular weight of depsipeptides inhibiting histone deacetylases in tumor cells.

Key

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

a

Bassianolide

Cyclo[N(Me)Leu-D-Oval-N(Me)Leu-D-Oval-N(Me)Leu-D-Oval-N(Me)Leu-D-Oval]

909.2

[55]

b

Clavatustide

Cyclo [2Abz-2Abz-D-Ophe-N(Et)Gly]

471.5

[56]

c

Cryptophycin

10-[(3-chloro-4-methoxyphenyl)methyl]-6-methyl-3-(2-methylpropyl)-16-[1-(3-phenyloxiran-2-yl)ethyl]-1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone

655.2

[57]

d

Largazole

S-[I-4-[(5R,8S,11S)-5-Me-6,9,13-trioxo-8-propan-2-yl-10-oxa-3,17-dithia-7,14,19,20-tetrazatriCyclo [14.2.1.12,5]icosa-1(18),2(20),16(19)-trien-11-yl]but-3-enyl] octanethioate

622.9

[58]

e

Lyngbyabellin

(7S,14S,18S)-7-[(2S)-butan-2-yl]-14-(4,4-dichloropentyl)-18-(2-hydroxypropan-2-yl)-15,15-dimethyl-13,17-dioxa-9,20-dithia-3,6,22,23-tetrazatriCyclo [17.2.1.18,11]tricosa-1(21),8(23),10,19(22)-tetraene-2,5,12,16-tetrone

691.7

[18]

f

Romidepsin

7-ethylidene-4,21-di(propan-2-yl)-2-oxa-12,13-dithia-5,8,20,23-tetrazabiCyclo [8.7.6]tricos-16-ene-3,6,9,19,22-pentone

540.7

[59]

g

Sansalvamide

Cyclo[L-leucyl-N-oxa-L-leucyl-L-valyl-L-leucyl-L-phenylalanyl]

586.8

[60]

2.5. Depsipeptides Disrupting Microfilaments

Microtubule–actin interactions are fundamental for many cellular processes such as cell motility, cell division, and cytoskeleton dynamics [61]. Many depsipeptides derived from bacteria exert activities against microfilaments, causing complete depolymerization of microtubules and hyper-polymerization of actin [62]. Moreover, they have shown the ability to destabilize microtubules, inducing caspase and Bcl-2 activation, eventually resulting in cell cycle arrest and apoptosis [63].
Here, the researchers explore the depsipeptides that disrupt microfilaments in tumor cells (Figure 6 and Table 5).
Figure 6. Models of depsipeptides inducing microfilament disruption in tumor cells. (a) Desmethoxymajusculamide C; (b) Dolastatin 10; (c) Miuraenamide; (d) Nobilamide I.
Table 5. Formula and molecular weight of depsipeptides inducing microfilament disruption in tumor cells.

Key

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

a

Desmethoxymajusculamide C

Cyclo[Ala-Unk-Ala-Unk-Gly-N(Me)Ile-Gly-N(Me)Val-N(Me)Phe]

955.2

[64]

b

Dolastatin

N(Me2)Val-Val-Unk

785.1

[65]

c

Miuraenamide

(3E,15E)-6-[(3-bromo-4-hydroxyphenyl)methyl]-3-[methoxy(phenyl)methylidene]-7,9,16,19-tetramethyl-1-oxa-4,7,10-triazacyclononadec-15-ene-2,5,8,11-tetrone

684.6

[66]

d

Nobilamide

Propionyl-D-Phe-D-Leu-Phe-D-aThr-Val-Ala-Abu(2,3-dehydro)-OH

836.0

[67]

2.6. Depsipeptides Inhibiting Cell Growth

The loss of normal cell cycle regulation is one of the cornerstones of human cancer emergence [68]. Thus, compounds that inhibit or terminate mitosis have been the emphasis of clinical trials for many years, with some achieving significant success [69]. Certain depsipeptides are of particular interest in this field [70][71], mainly skyllamycins and stereocalpin compounds that inhibit transcription factors and survival signaling [72][73].
Thus, the researchers delved into the depsipeptides that inhibit cell growth (Table 6 and Figure 7).
Figure 7. Models of skyllamycins (a) and stereocalpin (b) depsipeptides inhibiting the growth of tumor cells.
Table 6. Formula and molecular weight of depsipeptides inhibiting the growth of tumor cells.

Key

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

a

Skyllamycins

N-[I-3-[2-[(Z)-1-Propenyl]phenyl]propenoyl]-Cyclo[L-Thr*-L-Ala-[(3S)-3-Me-L-Asp-]-Gly-[(βS)-β-hydroxy-L-Phe-]-L-Pro-[(βS)-β-hydroxy-O-Me-L-Tyr-]-D-Trp-[(2S)-2-hydroxy-Gly-]-D-Leu-[(3S)-3-hydroxy-D-Leu-]-]

1483.6

[74]

b

Stereocalpin

Cyclo[Phe-N(Me)Phe-Unk]

492.6

[75]

2.7. Depsipeptides Inhibiting Topoisomerases

Topoisomerases are enzymes that relieve torsional stress within DNA during replication and transcription by cleaving one strand to unwind the supercoiled DNA [76][77]. Inhibiting these enzymes leads to DNA damage, nicotinamide adenine dinucleotide phosphate oxidase-dependent generation of reactive oxygen species, p21 activation, and eventual cell cycle arrest and senescence [78].
Here, the researchers describe the relevant depsipeptides targeting topoisomerases in cancer cells (Table 7 and Figure 8).
Figure 8. Models of fusaristatin A (a) and neo-N-methylsansalvamide (b) depsipeptides targeting topoisomerases in tumor cells.
Table 7. Formula and molecular weight of depsipeptides targeting topoisomerases in tumor cells.

Key

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

a

Fusaristatin A

3-[6,13-dimethyl-10-methylidene-2,5,9,12-tetraoxo-14-[(5E,7E)-3,7,11-trimethyl-4-oxoheptadeca-5,7-dienyl]-1-oxa-4,8,11-triazacyclotetradec-3-yl]propanamide

658.9

[79]

b

N-methylsansalvamide

Cyclo[Leu-Oleu-Val-N(Me)Leu-Phe]

600.8

[28]

2.8. Oncosis

Oncosis is a form of programmed cell death characterized by organelle swelling and membrane disruption [80]. It is a type of cell injury that involves energy depletion, impairment of ionic pumps, swelling, dilation of the Golgi apparatus and endoplasmic reticulum, chromatin clumping, and the formation of cytoplasmic blebs [81].
Compared with apoptosis, oncosis eventually results in necrosis with karyolysis and cell swelling, instead of karyorrhexis and cell shrinkage [82]. One of the most studied oncosis-inducing depsipeptides in cancer cells is kahalalide F (Figure 9 and Table 8), a molecule that causes disruption of the mitochondrial membrane potential and alters the permeability of lysosomal membranes [42].
Figure 9. Model of kahalalide F: the only depsipeptide inducing oncosis in tumor cells.
Table 8. Formula and molecular weight of kahalalide F, the only depsipeptide inducing oncosis in tumor cells.

Depsipeptide

IUPAC Formula

Molecular Weight

(g/mol)

Ref.

Kahalalide F

L-Val, N-(5-Me-1-oxohexyl)-D-valyl-l-threonyl-l-valyl-D-valyl-D-prolyl-l-ornithyl-D-alloisoleucyl-D-allothreonyl-D-alloisoleucyl-D-valyl-l-phenylalanyl-(2z)-2-amino-2-butenoyl-, (13->8)-lactone

1477.9

[83]

3. Current Methods for Purification and Synthesis of Depsipeptides

3.1. Proved Methods for Purifying Depsipeptides

Nowadays, depsipeptides can be purified using different methods [84][85]. When derived from natural sources, the organism-produced depsipeptides can be analyzed using high-performance liquid chromatography (HPLC) [86][87]. Some studies have coupled a photo-diode array and used ultra-performance liquid chromatography in tandem with high-resolution mass spectrometry [88].
Frequently, in vitro and even in vivo assays are performed simultaneously during purification to analyze the biological properties of interest [89]. Yu et al. used reversed-phase HPLC (RP-HPLC) to purify and quantify depsipeptides and short oligomers of glycine [86]. In 2006, Matsuo et al. also used RP-HPLC to purify urukthapelstatin [90], a depsipeptide with inhibitory effects on human lung cancer (A549) cells [87].
RP-HPLC was also used to purify trikoveramides A–C, peptides that belong to the kulolide superfamily and inhibited the growth of human leukemia cells (MOLT-4) [91]. The well-known anticancer peptide, coibamide A, was also purified by RP-HPLC [92]. In 2015, Kaur et al. achieved the purification of the novel cyclic depsipeptide, YM-280193, with the ability to inhibit platelet aggregation [93].

3.2. Synthesis of Depsipeptides

Depsipeptides were originally synthesized to improve the solubility of ultra-long peptides by adding an ester-linked residue that would alter the linear structure [94]. In addition to the presence of at least one ester and one amide bond in their structures, most depsipeptides contain a wide variety of acyl groups and moieties added through the assembly line [16].
Certain authors have proposed the synthesis of these peptides using solid-phase peptide synthesis with a fluorenylmethoxycarbonyl (Fmoc)-protected N-terminus, lysine, or aspartic acid as the charged entities, and lactic acid as the ester moiety to maintain hydrophobicity [85]. A stepwise Fmoc-based solid-phase methodology to create a highly complex depsipeptide has been reported, with the addition of small amounts of organic acids to improve this stepwise approach and minimize secondary reactions [95].
For instance, coibamide A has been synthesized using Fmoc-based solid-phase synthesis, but with the initial step being the attachment of the Fmoc-Tyr-allyl ester to the free hydroxyl group on a 2-chlorotrityl chloride resin [92]. Another depsipeptide, teixobactin, and its analogs have also been synthesized using Fmoc-based solid-phase synthesis [96].
A recent modification of the Fmoc-based solid-phase synthesis involved the use of ultrasonication methods, an intervention that saved both material and reaction time while improving the synthesis yield of complex peptide sequences [97]. A novel and efficient approach to generate depsipeptides emerged in the form of isocyanide-based consecutive Bargellini–Passerini multicomponent reactions [98].

4. Recognized Disadvantages of Depsipeptides

Even though depsipeptides are derived from natural resources, their commercialization requires synthetic production, which generates a vast amount of waste [99]. To tackle these issues, cyclization has been proposed to minimize dimerization and oligomerization, which in turn involves high dilutions of the peptide [100]. However, a synthetic route needs to be designed, optimized, and tailored to each particular depsipeptide, rendering a general synthetic method impossible [95].
Synthetic depsipeptides have to replicate the capability of modulating DNA methylation, acetylation, and histone methylation through specific biomolecules with unique structural characteristics [101], making this a complex, strenuous, and most likely, high-cost process. Furthermore, these peptides tend to be ionophoric in nature and require metal salt templates to be added to the macrocyclization reactions as they enhance the rate of cyclization [99]. Moreover, the cell-binding capacity of these peptides and the uncertain toxicity of their degradation products are some other concerns [102].

References

  1. Bray, F.; Laversanne, M.; Weiderpass, E.; Soerjomataram, I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 2021, 127, 3029–3030.
  2. World Health Organization. Global Health Estimates for 2020: Deaths by Cause, Age, Sex, by Country and by Region, 2000–2019; WHO: Geneva, Switzerland, 2020.
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA. Cancer J. Clin. 2021, 71, 209–249.
  4. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789.
  5. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33.
  6. Pilleron, S.; Sarfati, D.; Janssen-Heijnen, M.; Vignat, J.; Ferlay, J.; Bray, F.; Soerjomataram, I. Global cancer incidence in older adults, 2012 and 2035: A population-based study. Int. J. Cancer 2019, 144, 49–58.
  7. Pilleron, S.; Soto-Perez-de-Celis, E.; Vignat, J.; Ferlay, J.; Soerjomataram, I.; Bray, F.; Sarfati, D. Estimated global cancer incidence in the oldest adults in 2018 and projections to 2050. Int. J. Cancer 2021, 148, 601–608.
  8. Trinidad-Calderón, P.A.; López-Castillo, L.-M.; Gallegos-Martínez, S.; Trujillo-de-Santiago, G.; García-Lara, S.; Álvarez, M.M. nurP28, a New-to-Nature Zein-Derived Peptide, Enhances the Therapeutic Effect of Docetaxel in Breast Cancer Monolayers and Spheroids. Molecules 2022, 27, 2824.
  9. Serna-Thome, G.; Castro-Eguiluz, D.; Fuchs-Tarlovsky, V.; Sanchez-Lopez, M.; Delgado-Olivares, L.; Coronel-Martinez, J.; Molina-Trinidad, E.M.; De La Torre, M.; Cetina-Perez, L. Use of functional foods and oral supplements as adjuvants in cancer treatment. Rev. Investig. Clin. 2018, 70, 136–146.
  10. Mondal, A.; Bose, S.; Banerjee, S.; Patra, J.K.; Malik, J.; Mandal, S.K.; Kilpatrick, K.L.; Das, G.; Kerry, R.G.; Fimognari, C.; et al. Marine Cyanobacteria and Microalgae Metabolites—A Rich Source of Potential Anticancer Drugs. Mar. Drugs 2020, 18, 476.
  11. Sharma, P.; Kaur, H.; Kehinde, B.A.; Chhikara, N.; Sharma, D.; Panghal, A. Food-Derived Anticancer Peptides: A Review. Int. J. Pept. Res. Ther. 2021, 27, 55–70.
  12. Zhang, Q.T.; Liu, Z.D.; Wang, Z.; Wang, T.; Wang, N.; Wang, N.; Zhang, B.; Zhao, Y.F. Recent advances in small peptides of marine origin in cancer therapy. Mar. Drugs 2021, 19, 115.
  13. Zhang, J.N.; Xia, Y.X.; Zhang, H.J. Natural cyclopeptides as anticancer agents in the last 20 years. Int. J. Mol. Sci. 2021, 22, 3973.
  14. Adrover-Castellano, M.L.; Schmidt, J.J.; Sherman, D.H. Biosynthetic Cyclization Catalysts for the Assembly of Peptide and Polyketide Natural Products. ChemCatChem 2021, 13, 2095–2116.
  15. Taevernier, L.; Wynendaele, E.; Gevaert, B.; Spiegeleer, B. Chemical Classification of Cyclic Depsipeptides. Curr. Protein Pept. Sci. 2017, 18, 425–452.
  16. Alonzo, D.A.; Schmeing, T.M. Biosynthesis of depsipeptides, or Depsi: The peptides with varied generations. Protein Sci. 2020, 29, 2316–2347.
  17. Rangel, M.; Santana, C.; Pinheiro, A.; Anjos, L.; Barth, T.; Júnior, O.; Fontes, W.; Castro, M. Marine Depsipeptides as Promising Pharmacotherapeutic Agents. Curr. Protein Pept. Sci. 2016, 18, 72–91.
  18. Lee, Y.; Phat, C.; Hong, S.C. Structural diversity of marine cyclic peptides and their molecular mechanisms for anticancer, antibacterial, antifungal, and other clinical applications. Peptides 2017, 95, 94–105.
  19. Pavlicevic, M.; Maestri, E.; Marmiroli, M. Marine Bioactive Peptides—An Overview of Generation, Structure and Application with a Focus on Food Sources. Mar. Drugs 2020, 18, 424.
  20. Khalifa, S.A.M.; Elias, N.; Farag, M.A.; Chen, L.; Saeed, A.; Hegazy, M.E.F.; Moustafa, M.S.; El-Wahed, A.A.; Al-Mousawi, S.M.; Musharraf, S.G.; et al. Marine natural products: A source of novel anticancer drugs. Mar. Drugs 2019, 17, 491.
  21. Kang, H.; Choi, M.-C.; Seo, C.; Park, Y. Therapeutic Properties and Biological Benefits of Marine-Derived Anticancer Peptides. Int. J. Mol. Sci. 2018, 19, 919.
  22. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592.
  23. Urbańska, K.; Orzechowski, A. Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int. J. Mol. Sci. 2019, 20, 2085.
  24. Pistritto, G.; Trisciuoglio, D.; Ceci, C.; Garufi, A.; D’Orazi, G. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging 2016, 8, 603–619.
  25. Giampazolias, E.; Tait, S.W.G. Mitochondria and the hallmarks of cancer. FEBS J. 2016, 283, 803–814.
  26. Sharma, A.; Boise, L.H.; Shanmugam, M. Cancer metabolism and the evasion of apoptotic cell death. Cancers 2019, 11, 1144.
  27. Moschos, M.M.; Dettoraki, M.; Androudi, S.; Kalogeropoulos, D.; Lavaris, A.; Garmpis, N.; Damaskos, C.; Garmpi, A.; Tsatsos, M. The role of histone deacetylase inhibitors in uveal melanoma: Current evidence. Anticancer Res. 2018, 38, 3817–3824.
  28. Lee, H.S.; Phat, C.; Choi, S.U.; Lee, C. Synergistic effect of a novel cyclic pentadepsipeptide, neoN-methylsansalvamide, and paclitaxel on human multidrug resistance cancer cell lines. Anticancer Drugs 2013, 24, 455–460.
  29. Wan, X.; Serrill, J.D.; Humphreys, I.R.; Tan, M.; McPhail, K.L.; Ganley, I.G.; Ishmael, J.E. ATG5 promotes death signaling in response to the cyclic depsipeptides coibamide a and apratoxin A. Mar. Drugs 2018, 16, 77.
  30. Okada, K.; Hakata, S.; Terashima, J.; Gamou, T.; Habano, W.; Ozawa, S. Combination of the histone deacetylase inhibitor depsipeptide and 5-fluorouracil upregulates major histocompatibility complex class II and p21 genes and activates caspase-3/7 in human colon cancer HCT-116 cells. Oncol. Rep. 2016, 36, 1875–1885.
  31. Cai, W.; Ratnayake, R.; Gerber, M.H.; Chen, Q.-Y.; Yu, Y.; Derendorf, H.; Trevino, J.G.; Luesch, H. Development of apratoxin S10 (Apra S10) as an anti-pancreatic cancer agent and its preliminary evaluation in an orthotopic patient-derived xenograft (PDX) model. Investig. New Drugs 2019, 37, 364–374.
  32. Caloni, F.; Fossati, P.; Anadón, A.; Bertero, A. Beauvericin: The beauty and the beast. Environ. Toxicol. Pharmacol. 2020, 75, 103349.
  33. He, W.; Qiu, H.-B.; Chen, Y.-J.; Xi, J.; Yao, Z.-J. Total synthesis of proposed structure of coibamide A, a highly N- and O-methylated cytotoxic marine cyclodepsipeptide. Tetrahedron Lett. 2014, 55, 6109–6112.
  34. Zuo, W.; Kwok, H.F. Development of marine-derived compounds for cancer therapy. Mar. Drugs 2021, 19, 342.
  35. Taevernier, L.; Veryser, L.; Roche, N.; Peremans, K.; Burvenich, C.; Delesalle, C.; De Spiegeleer, B. Human skin permeation of emerging mycotoxins (beauvericin and enniatins). J. Expo. Sci. Environ. Epidemiol. 2016, 26, 277–287.
  36. Kwan, J.C.; Rocca, J.R.; Abboud, K.A.; Paul, V.J.; Luesch, H. Total structure determination of grassypetolide, a new marine cyanobacterical cytotoxin. Org. Lett. 2008, 10, 789–792.
  37. Xue, Y.; Zhao, P.; Quan, C.; Zhao, Z.; Gao, W.; Li, J.; Zu, X.; Fu, D.; Feng, S.; Bai, X.; et al. Cyanobacteria-derived peptide antibiotics discovered since 2000. Peptides 2018, 107, 17–24.
  38. Huang, W.; Ren, R.G.; Dong, H.Q.; Wei, B.G.; Lin, G.Q. Diverse synthesis of marine cyclic depsipeptide lagunamide A and its analogues. J. Org. Chem. 2013, 78, 10747–10762.
  39. Levert, A.; Alvariño, R.; Bornancin, L.; Abou Mansour, E.; Burja, A.M.; Genevière, A.M.; Bonnard, I.; Alonso, E.; Botana, L.; Banaigs, B. Structures and Activities of Tiahuramides A-C, Cyclic Depsipeptides from a Tahitian Collection of the Marine Cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2018, 81, 1301–1310.
  40. Bialik, S.; Dasari, S.K.; Kimchi, A. Autophagy-dependent cell death—Where, how and why a cell eats itself to death. J. Cell Sci. 2018, 131, jcs215152.
  41. Zhang, L.; Wang, Y.; Huang, W.; Wei, Y.; Jiang, Z.; Kong, L.; Wu, A.A.; Hu, Z.; Huang, H.; Xu, Q.; et al. Biosynthesis and Chemical Diversification of Verucopeptin Leads to Structural and Functional Versatility. Org. Lett. 2020, 22, 4366–4371.
  42. Kitagaki, J.; Shi, G.; Miyauchi, S.; Murakami, S.; Yang, Y. Cyclic depsipeptides as potential cancer therapeutics. Anticancer Drugs 2015, 26, 259–271.
  43. Liu, J.; Zhu, X.; Kim, S.J.; Zhang, W. Antimycin-type depsipeptides: Discovery, biosynthesis, chemical synthesis, and bioactivities. Nat. Prod. Rep. 2016, 33, 1146–1165.
  44. Urbaniak, M.; Stępień, Ł.; Uhlig, S. Evidence for naturally produced beauvericins containing N-Methyl-Tyrosine in Hypocreales Fungi. Toxins 2019, 11, 182.
  45. Urbaniak, M.; Waskiewicz, A.; Stepien, Ł. Fusarium cyclodepsipeptide mycotoxins: Chemistry, biosynthesis, and occurrence. Toxins 2020, 12, 765.
  46. Tian, J.; Han, J.J.; Zhang, X.; He, L.W.; Zhang, Y.J.; Bao, L.; Liu, H.W. New Cyclohexadepsipeptides from an Entomogenous Fungus Fusarium proliferatum and Their Cytotoxicity and Autophagy-Inducing Activity. Chem. Biodivers. 2016, 13, 852–860.
  47. Bunyapaiboonsri, T.; Vongvilai, P.; Auncharoen, P.; Isaka, M. Cyclohexadepsipeptides from the Filamentous Fungus Acremonium sp. BCC 2629. Helv. Chim. Acta 2012, 95, 963–972.
  48. Marinaccio, L.; Stefanucci, A.; Scioli, G.; Della Valle, A.; Zengin, G.; Cichelli, A.; Mollica, A. Peptide Human Neutrophil Elastase Inhibitors from Natural Sources: An Overview. Int. J. Mol. Sci. 2022, 23, 2924.
  49. Al-Awadhi, F.H.; Paul, V.J.; Luesch, H. Structural Diversity and Anticancer Activity of Marine-Derived Elastase Inhibitors: Key Features and Mechanisms Mediating the Antimetastatic Effects in Invasive Breast Cancer. ChemBioChem 2018, 19, 815–825.
  50. Chen, Q.-Y.; Luo, D.; Seabra, G.M.; Luesch, H. Ahp-Cyclodepsipeptides as tunable inhibitors of human neutrophil elastase and kallikrein 7: Total synthesis of tutuilamide A, serine protease selectivity profile and comparison with lyngbyastatin 7. Bioorg. Med. Chem. 2020, 28, 115756.
  51. Geada, P.; Gkelis, S.; Teixeira, J.; Vasconcelos, V.; Vicente, A.A.; Fernandes, B. Cyanobacterial Toxins as a High Value-Added Product; Elsevier Ltd.: Amsterdam, The Netherlands, 2017; ISBN 9780081010273.
  52. Favas, R.; Morone, J.; Martins, R.; Vasconcelos, V.; Lopes, G. Cyanobacteria and microalgae bioactive compounds in skin-ageing: Potential to restore extracellular matrix filling and overcome hyperpigmentation. J. Enzyme Inhib. Med. Chem. 2021, 36, 1829–1838.
  53. Zhang, H.; Shang, Y.P.; Chen, H.Y.; Li, J. Histone deacetylases function as novel potential therapeutic targets for cancer. Hepatol. Res. 2017, 47, 149–159.
  54. Garmpis, N.; Damaskos, C.; Garmpi, A.; Dimitroulis, D.; Spartalis, E.; Margonis, G.A.; Schizas, D.; Deskou, I.; Doula, C.; Magkouti, E.; et al. Targeting histone deacetylases in malignant melanoma: A future therapeutic agent or just great expectations? Anticancer Res. 2017, 37, 5355–5362.
  55. Mun, B.; Park, Y.J.; Sung, G.H.; Lee, Y.; Kim, K.H. Synthesis and antitumor activity of (−)-bassianolide in MDA-MB 231 breast cancer cells through cell cycle arrest. Bioorg. Chem. 2016, 69, 64–70.
  56. Chettu, S.K.; Madhu, R.B.; Raolji, G.B.; Babu, K.R.; Rao, N.S.K.; Gopalakrishnan, S.; Ismail, A.; Reddy, G.B.; Shafi, S. First total synthesis of cyclodepsipeptides clavatustide A and B and their enantiomers. RSC Adv. 2016, 6, 61555–61565.
  57. Schmidt, J.J.; Khatri, Y.; Brody, S.I.; Zhu, C.; Pietraszkiewicz, H.; Valeriote, F.A.; Sherman, D.H. A versatile chemoenzymatic synthesis for the discovery of potent cryptophycin analogs. ACS Chem. Biol. 2020, 15, 524–532.
  58. Poli, G.; Di Fabio, R.; Ferrante, L.; Summa, V.; Botta, M. Largazole Analogues as Histone Deacetylase Inhibitors and Anticancer Agents: An Overview of Structure–Activity Relationships. ChemMedChem 2017, 12, 1917–1926.
  59. Smolewski, P.; Robak, T. The discovery and development of romidepsin for the treatment of T-cell lymphoma. Expert Opin. Drug Discov. 2017, 12, 859–873.
  60. Romans-Fuertes, P.; Sondergaard, T.E.; Sandmann, M.I.H.; Wollenberg, R.D.; Nielsen, K.F.; Hansen, F.T.; Giese, H.; Brodersen, D.E.; Sørensen, J.L. Identification of the non-ribosomal peptide synthetase responsible for biosynthesis of the potential anti-cancer drug sansalvamide in FusariumFusarium solani. Curr. Genet. 2016, 62, 799–807.
  61. Kigoshi, H.; Kita, M. Antitumor Effects of Sea Hare-Derived Compounds in Cancer. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 701–739. ISBN 9783319071459.
  62. Tan, L.T.; Gupta, D.K. Molecular Targets of Anticancer Agents from Filamentous Marine Cyanobacteria. In Handbook of Anticancer Drugs from Marine Origin; Springer International Publishing: Cham, Switzerland, 2015; pp. 571–592. ISBN 9783319071459.
  63. Mandal, S.; Rath, J. Anticancer Drug Development from Cyanobacteria. In Extremophilic Cyanobacteria for Novel Drug Development. SpringerBriefs in Pharmaceutical Science & Drug Development; Springer: Cham, Switzerland, 2015; pp. 63–78.
  64. Śliżewska, A.; Żymańczyk-Duda, E. Cyanobacteria as valuable tool in biotechnology. Catalysts 2021, 11, 1259.
  65. Zhou, W.; Nie, X.D.; Zhang, Y.; Si, C.M.; Zhou, Z.; Sun, X.; Wei, B.G. A practical approach to asymmetric synthesis of dolastatin 10. Org. Biomol. Chem. 2017, 15, 6119–6131.
  66. Tost, M.; Andler, O.; Kazmaier, U. A Matteson Homologation-Based Synthesis of Doliculide and Derivatives. Eur. J. Org. Chem. 2021, 2021, 6459–6471.
  67. Yamashita, T.; Matoba, H.; Kuranaga, T.; Inoue, M. Total syntheses of nobilamides B and D: Application of traceless Staudinger ligation. Tetrahedron 2014, 70, 7746–7752.
  68. Dominguez-Brauer, C.; Thu, K.L.; Mason, J.M.; Blaser, H.; Bray, M.R.; Mak, T.W. Targeting Mitosis in Cancer: Emerging Strategies. Mol. Cell 2015, 60, 524–536.
  69. Haschka, M.; Karbon, G.; Fava, L.L.; Villunger, A. Perturbing mitosis for anti-cancer therapy: Is cell death the only answer? EMBO Rep. 2018, 19, e45440.
  70. Mehta, A.; Soni, V.K.; Shukla, D.; Vishvakarma, N.K. Cyanobacteria: A Potential Source of Anticancer Drugs; Academic Press: Cambridge, MA, USA, 2020; ISBN 9780128193112.
  71. Ahmed, S.; Khan, H.; Fakhri, S.; Aschner, M.; Cheang, W.S. Therapeutic potential of marine peptides in cervical and ovarian cancers. Mol. Cell. Biochem. 2022, 477, 605–619.
  72. Zhu, J.; Zhang, S.; Zechel, D.L.; Paululat, T.; Bechthold, A. Rational Design of Hybrid Natural Products by Utilizing the Promiscuity of an Amide Synthetase. ACS Chem. Biol. 2019, 14, 1793–1801.
  73. Byeon, H.E.; Park, B.K.; Yim, J.H.; Lee, H.K.; Moon, E.Y.; Rhee, D.K.; Pyo, S. Stereocalpin A inhibits the expression of adhesion molecules in activated vascular smooth muscle cells. Int. Immunopharmacol. 2012, 12, 315–325.
  74. Giltrap, A.M.; Haeckl, F.P.J.; Kurita, K.L.; Linington, R.G.; Payne, R.J. Synthetic Studies Toward the Skyllamycins: Total Synthesis and Generation of Simplified Analogues. J. Org. Chem. 2018, 83, 7250–7270.
  75. Cimmino, A.; Nimis, P.L.; Masi, M.; De Gara, L.; van Otterlo, W.A.L.; Kiss, R.; Evidente, A.; Lefranc, F. Have lichenized fungi delivered promising anticancer small molecules? Phytochem. Rev. 2019, 18, 1–36.
  76. Murai, J.; Thomas, A.; Miettinen, M.; Pommier, Y. Schlafen 11 (SLFN11), a restriction factor for replicative stress induced by DNA-targeting anti-cancer therapies. Pharmacol. Ther. 2019, 201, 94–102.
  77. Mohan, C.D.; Rangappa, S.; Nayak, S.C.; Jadimurthy, R.; Wang, L.; Sethi, G.; Garg, M.; Rangappa, K.S. Bacteria as a treasure house of secondary metabolites with anticancer potential. Semin. Cancer Biol. 2021, 86, 998–1013.
  78. Watters, D.J. Ascidian toxins with potential for drug development. Mar. Drugs 2018, 16, 162.
  79. Hegge, A.; Lønborg, R.; Nielsen, D.M.; Sørensen, J.L. Factors influencing production of fusaristatin A in Fusarium graminearum. Metabolites 2015, 5, 184–191.
  80. Puig, B.; Brenna, S.; Magnus, T. Molecular communication of a dying neuron in stroke. Int. J. Mol. Sci. 2018, 19, 2834.
  81. Weerasinghe, P.; Hallock, S.; Brown, R.; Buja, L.M. Oncosis. In Apoptosis and Beyond; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2018; pp. 567–582.
  82. Tsai, C.M. AqF026 may act as a cancer therapeutic agent via inducing cancer cell oncosis. Med. Hypotheses 2020, 140, 109685.
  83. Dayanidhi, D.L.; Thomas, B.C.; Osterberg, J.S.; Vuong, M.; Vargas, G.; Kwartler, S.K.; Schmaltz, E.; Dunphy-Daly, M.M.; Schultz, T.F.; Rittschof, D.; et al. Exploring the Diversity of the Marine Environment for New Anti-cancer Compounds. Front. Mar. Sci. 2021, 7, 614766.
  84. Kaneda, M.; Sueyoshi, K.; Teruya, T.; Ohno, H.; Fujii, N.; Oishi, S. Total synthesis of odoamide, a novel cyclic depsipeptide, from an Okinawan marine cyanobacterium. Org. Biomol. Chem. 2016, 14, 9093–9104.
  85. Nguyen, M.M.; Ong, N.; Suggs, L. A general solid phase method for the synthesis of depsipeptides. Org. Biomol. Chem. 2013, 11, 1167–1170.
  86. Campbell, T.D.; Febrian, R.; Kleinschmidt, H.E.; Smith, K.A.; Bracher, P.J. Quantitative Analysis of Glycine Oligomerization by Ion-Pair Chromatography. ACS Omega 2019, 4, 12745–12752.
  87. Phyo, Y.; Ribeiro, J.; Fernandes, C.; Kijjoa, A.; Pinto, M.M.M. Marine natural peptides: Determination of absolute configuration using liquid chromatography methods and evaluation of bioactivities. Molecules 2018, 23, 306.
  88. Greco, C.; Pfannenstiel, B.T.; Liu, J.C.; Keller, N.P. Depsipeptide Aspergillicins Revealed by Chromatin Reader Protein Deletion. ACS Chem. Biol. 2019, 14, 1121–1128.
  89. Arumugam, V.; Venkatesan, M.; Ramachandran, S.; Sundaresan, U. Bioactive Peptides from Marine Ascidians and Future Drug Development–A Review. Int. J. Pept. Res. Ther. 2018, 24, 13–18.
  90. Matsuo, Y.; Kanoh, K.; Imagawa, H.; Adachi, K.; Nishizawa, M.; Shizuri, Y. Urukthapelstatin A, a novel cytotoxic substance from marine-derived Mechercharimyces asporophorigenens YM11-542: II. Physico-chemical properties and structural elucidation. J. Antibiot. 2007, 60, 256–260.
  91. Phyo, M.Y.; Katermeran, N.P.; Goh, J.X.; Tan, L.T. Trikoveramides A-C, cyclic depsipeptides from the marine cyanobacterium Symploca hydnoides. Phytochemistry 2021, 190, 112879.
  92. Sable, G.A.; Park, J.; Kim, H.; Lim, S.J.; Jang, S.; Lim, D. Solid-Phase Total Synthesis of the Proposed Structure of Coibamide A and Its Derivative: Highly Methylated Cyclic Depsipeptides. Eur. J. Org. Chem. 2015, 2015, 7043–7052.
  93. Kaur, H.; Harris, P.W.R.; Little, P.J.; Brimble, M.A. Total synthesis of the cyclic depsipeptide YM-280193, a platelet aggregation inhibitor. Org. Lett. 2015, 17, 492–495.
  94. Pieszka, M.; Sobota, A.M.; Gačanin, J.; Weil, T.; Ng, D.Y.W. Orthogonally Stimulated Assembly/Disassembly of Depsipeptides by Rational Chemical Design. ChemBioChem 2019, 20, 1376–1381.
  95. Lobo-Ruiz, A.; Tulla-Puche, J. General Fmoc-Based Solid-Phase Synthesis of Complex Depsipeptides Circumventing Problematic Fmoc Removal. Eur. J. Org. Chem. 2020, 2020, 183–192.
  96. Jin, K.; Sam, I.H.; Po, K.H.L.; Lin, D.; Ghazvini Zadeh, E.H.; Chen, S.; Yuan, Y.; Li, X. Total synthesis of teixobactin. Nat. Commun. 2016, 7, 12394.
  97. Merlino, F.; Tomassi, S.; Yousif, A.M.; Messere, A.; Marinelli, L.; Grieco, P.; Novellino, E.; Cosconati, S.; Di Maro, S. Boosting Fmoc Solid-Phase Peptide Synthesis by Ultrasonication. Org. Lett. 2019, 21, 6378–6382.
  98. Farhid, H.; Rostami, M.M.; Shaabani, A.; Notash, B. Synthesis of Depsipeptides via Isocyanide-Based Consecutive Bargellini-Passerini Multicomponent Reactions. SynOpen 2021, 5, 167–172.
  99. Batiste, S.M.; Johnston, J.N. Evidence for Ion-Templation during Macrocyclooligomerization of Depsipeptides. J. Am. Chem. Soc. 2018, 140, 4560–4568.
  100. Jing, X.; Jin, K. A gold mine for drug discovery: Strategies to develop cyclic peptides into therapies. Med. Res. Rev. 2020, 40, 753–810.
  101. Conte, M.; Fontana, E.; Nebbioso, A.; Altucci, L. Marine-derived secondary metabolites as promising epigenetic bio-compounds for anticancer therapy. Mar. Drugs 2021, 19, 15.
  102. Eckes, K.M.; Baek, K.; Suggs, L.J. Design and Evaluation of Short Self-Assembling Depsipeptides as Bioactive and Biodegradable Hydrogels. ACS Omega 2018, 3, 1635–1644.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 265
Revisions: 3 times (View History)
Update Date: 10 Aug 2023
1000/1000
Video Production Service