Next Article in Journal
Zingerone Targets Status Epilepticus by Blocking Hippocampal Neurodegeneration via Regulation of Redox Imbalance, Inflammation and Apoptosis
Next Article in Special Issue
Sildenafil 4.0—Integrated Synthetic Chemistry, Formulation and Analytical Strategies Effecting Immense Therapeutic and Societal Impact in the Fourth Industrial Era
Previous Article in Journal
Novel Potent and Selective DPP-4 Inhibitors: Design, Synthesis and Molecular Docking Study of Dihydropyrimidine Phthalimide Hybrids
Previous Article in Special Issue
Is There a Role for the Therapeutic Drug Monitoring of Colistin? An Overview
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

2020 FDA TIDES (Peptides and Oligonucleotides) Harvest

by
Othman Al Musaimi
1,†,
Danah Al Shaer
2,†,
Fernando Albericio
1,3,4,* and
Beatriz G. de la Torre
2,*
1
School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
2
KRISP, School of Laboratory of Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
3
Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
4
Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2021, 14(2), 145; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020145
Submission received: 31 January 2021 / Revised: 9 February 2021 / Accepted: 9 February 2021 / Published: 11 February 2021
(This article belongs to the Special Issue The Story of Successful Drugs and Recent FDA-Approved Molecules)

Abstract

:
2020 has been an extremely difficult and challenging year as a result of the coronavirus disease 2019 (COVID-19) pandemic and one in which most efforts have been channeled into tackling the global health crisis. The US Food and Drug Administration (FDA) has approved 53 new drug entities, six of which fall in the peptides and oligonucleotides (TIDES) category. The number of authorizations for these kinds of drugs has been similar to that of previous years, thereby reflecting the consolidation of the TIDES market. Here, the TIDES approved in 2020 are analyzed in terms of chemical structure, medical target, mode of action, and adverse effects.

1. Introduction

In 2020, the Food and Drug Administration (FDA) has approved 53 drugs [1,2], giving a total of 228 entities (new chemical entities and biologics) authorized during the last 5 years (2016–2020) (Figure 1) [2]. Of the 53 drugs accepted in 2020, six are peptides and oligonucleotides (TIDES). In this regard, the last five years have witnessed the authorization of 23 TIDES. These numbers further strengthen the TIDES market, which, in number of drugs, accounts for around 10% of the total.
Although 2020 can be considered an excellent year from the perspective of both the total number of drugs approved and TIDES, it has been marked by the coronavirus disease 2019 (COVID-19) pandemic. However, this unique situation has allowed confirmation of the robustness of all pharmaceutical industry stakeholders. Indeed, in the space of approximately 10 months, the disease was identified and the first vaccinations were rolled out. From the perspective of TIDES, it is important to highlight that the three first vaccines approved by the regulatory agencies could be considered first-in-class and are based on either RNA or DNA. The explosion of oligonucleotides as drugs in recent years and later as key components of vaccines has been driven by intense research and investment in these intriguing molecules by many academic and industrial groups over more than 30 years.
The four peptides and two oligonucleotides approved by the FDA in 2020 are shown in Table 1, together with the indication, target, and route of administration.

2. Oligonucleotides

Oligonucleotides are becoming consolidated as therapeutic drugs after the success of this family in the treatment of various severe genetic disorders, mainly in the past few years. By the end of 2020, a dozen oligonucleotide drugs were on the market, nine of them having received approval since 2016. A summary of all authorized oligonucleotide drugs is shown in Table 2.

2.1. Viltolarsen (ViltepsoTM)

Viltolarsen is an antisense oligonucleotide consisting of a 21 phosphorodiamidate morpholino oligomer (PMO). In this kind of oligonucleotide analog, the natural sugar-phosphate backbone has been replaced by phosphorodiamidate-morpholino units, leading to a neutral backbone. The sequence of the bases is shown in (Figure 2) [4,5].
This drug is prescribed to treat Duchenne’s muscular dystrophy (DMD) amenable to exon 53 skipping [5]. DMD is an X chromosome-linked genetic disorder in which young patients suffer progressive deterioration of body muscles that ends with death in the second or third decade of life [4]. The dystrophin gene consists of 79 exons that encode for the 427-kDa cytoskeletal protein dystrophin. The protein protects and stabilizes the plasma membrane of muscle cells from the mechanical stress placed on muscle fibers upon contraction [4,6]. The absence of dystrophin damages muscle cell membranes, thereby leading to muscle degeneration and wasting [7]. Various genetic mutations in one or more of the exons mainly cause out-of-frame translational reading, which prevents transcription and consequently causes complete loss of the protein [4,5].
ViltepsoTM binds to exon 53 of the dystrophin mRNA precursor, thereby masking the exon. It then allows restoration of the reading frame. The reading process proceeds, but skipping exon 53 and consequently producing a shorter, but still functional, dystrophin protein [4,5]. This outcome delays muscle deterioration and turns the overall condition into a Becker muscular dystrophy (BMD)-like condition, thus increasing the life expectancy of DMD patients. [4].
ViltepsoTM is the third PMO antisense oligonucleotide used to treat DMD by exon skipping. The first two were eteplirsin and golodirsin for exons 51 and 53, respectively [6,8]. Sharing the same structure as viltolarsen but with an additional four subunits and a triethylene glycol moiety at its 5′ end, golodirsin reached the market in 2019. Exon 51 skipping therapy is applicable to 14% of DMD patients, while exon 53 skipping is applicable to 10%. A multi-exon skipping approach is now under investigation and is expected to be beneficial for 45% of DMD patients [4].
ViltepsoTM is administered intravenously and has shown some adverse effects, including cough, nasopharyngitis, upper respiratory tract infection, vomiting, and diarrhea [5,9]. Developed in Japan by Nippon Shinyaku and in collaboration with the National Center of Neurology and Psychiatry (NCNP), it was first approved in Japan in March 2020 [5] and received authorization from the FDA on 12 August of the same year [10].

2.2. Lumasiran (OxlumoTM)

Lumasiran is a double-stranded small interference RNA (siRNA). It consists of the sodium salt of a sense and an antisense strands containing some chemically modified units. Moreover, an N-acetyl galactosamine-bearing dendrimer is covalently attached to the 3′ terminus of the sense strand (Figure 3) [11]. After givosiran, which is used for the treatment of acute hepatic porphyria (AHP) and was approved in November 2019 [12], lumasiran is the second drug to exploit enhanced stabilization chemistry (ECT)-GalNAc conjugate technology [12,13].
Lumasiran is the first siRNA drug to be used for the treatment of primary hyperoxaluria type 1 (PH1), a hereditary disorder that results in the recurrent formation of kidney and bladder stones as a result of the deposition of endogenous oxalate as the poorly soluble calcium oxalate salt (CaOx) [14]. All previous treatments addressed mainly the symptoms rather than the primary cause [15]. The metabolism of hydroxyproline amino acid produces glyoxylate, which is further metabolized by certain hepatic enzymes to produce oxalate as final metabolite [16]. Oxalate is normally excreted in urine through the kidneys. Some genetic mutations cause a decrease in the production or activity of alanine-glyoxylate amino transferase (AGT), the enzyme responsible for breaking down glyoxylate. As a result, glyoxylate, and consequently the oxalate, accumulates [15,16,17]. Lumasiran (driven by the galactosamine moieties) targets hepatic cells and silences the gene encoding for glycolate oxidase (GO) enzyme, thereby halting its production. The depletion of this enzyme reduces the amounts of glyoxylate present and therefore the production of the oxalate [17].
The drug is administered subcutaneously and its main adverse effect is injection site reactions [11]. It was developed by Alnylam pharma and was approved by the FDA on 23 November 2020 [18].

3. Peptides

The last five years (2016–2020) have witnessed the authorization of 14 peptides, four being approved this year. These numbers reinforce the importance of peptides in the overall pharmaceutical market. Two of the class of 2020 are radiopharmaceuticals (64Cu and 68Ga), one a disulfide-containing peptide, and the last one the payload of an antibody drug conjugate (ADC). Table 3 shows the peptides approved by the FDA in the period 2016–2020.

3.1. Setmelanotide (ImcivreeTM)

Setmelanotide (formerly known as RM-493 or BIM-22493) is an eight-amino acid peptide that forms a cycle through a disulfide bridge between two Cys residues (Figure 4) [19]. The two D-amino acids (red) present in the sequence and its cyclic structure enhance resistance to proteolytic degradation, thereby increasing the half-life of the drug [20,21]. Furthermore, the positively charged amino acids (blue) are considered essential for ligand-receptor recognition and for activity [22].
Setmelanotide is prescribed for the treatment of rare genetic diseases of obesity caused by certain variants of the genes encoding for pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) all of which cause a deficiency of these molecules [23]. Patients with such genetic alterations suffer from excessive or extreme hunger, severe early-onset obesity, and endocrine disorders [24,25].
Setmelanotide acts as a selective melanocortin-4 (MC4) receptor agonist and presents a 20-fold higher potency than the endogenous agonist α-MSH [19,26]. In the brain, MC4 receptors are involved in regulating hunger and satiety, in addition to energy expenditure [27]. Deficiency of POMC, PCSK1, and LEPR cause the MC4 receptor pathway to be insufficiently activated. Therefore, the binding of setmelanotide to the MC4 receptor reactivates the pathway, inducing a reduction of hunger that promotes weight loss due to decreased caloric intake and increased energy expenditure. After receiving setmelanotide, a noticeable reduction in body weight, as well as reversing hyperphagia, is observed within 45 to 61 weeks [22,28].
Setmelanotide shows high efficacy with fewer side effects, especially those related to hypertension and adverse cardiovascular effects, which were observed in the first generation MC4R agonists, such as LY2112688 [22,27,29].
It is administered subcutaneously and common adverse effects include skin hyperpigmentation, nausea, headache, diarrhea, abdominal pain, back pain, fatigue, vomiting, depression, upper respiratory tract infection, and spontaneous penile erection [23]. It also has several warnings and precautions, such as disturbance of sexual arousal, depression and suicidal ideation, skin pigmentation and darkening of pre-existing nevi, risk of serious adverse reactions in neonates, and low birth weight infants due to benzyl alcohol preservative [23]. It was developed by Rhythm Pharmaceuticals and was approved by the FDA on 25 November 2020 [30].

3.2. 64Cu -DOTATATE (DetectnetTM)

64Cu -DOTATATE is composed of the radionuclide 64Cu (purple) chelated by DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) (blue), which is linked to the somatostatin analog octreotate peptide (black) (Figure 5). It is a radioactive diagnostic agent to be used with positron emission tomography (PET) for the localization of somatostatin receptor-positive neuroendocrine tumors (NETs) in adults [31]. Although 64Cu was previously used in nuclear medicine as 64Cu-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM) for PET imaging in head and neck cancer [32], and hypoxic myocardium [33], and in 64Cu-labeled anti-colorectal carcinoma mAbs (MAb 1A3) [34], 64Cu-DOTATATE is considered the first FDA-approved 64Cu-labeled radiopharmaceutical for Positron emission tomography–computed tomography (PET/CT) imaging.
Among the most important Cu radioisotopes, 64Cu is considered the most convenient for clinical applications since it has an intermediate half-life of 12.7 h and can decay via various routes, including electron capture, beta emission, and positron emission. Its low positron energy allows high quality and high-resolution images, plus the absence of abundant gamma emission (in contrast to other positron emitters), a type of emission that may also impair the imaging process [34]].
64Cu-DOTATATE outperforms 68Ga-DOTATOC, which was approved in 2019 [8]. This is explained by the fact that the radionuclide of 68Ga-DOTATOC has a shorter half-life (68 min), and its images are of lower resolution as a result of its higher positron energy [8]. A study by Johnbeck et al. showed that 64Cu-DOTATATE gave improved and greater detection of lesions than 68Ga-DOTATOC [35]. Furthermore, another study carried out by Pfeifer et al. also demonstrated a higher spatial resolution of the images taken by 64Cu- DOTATATE versus those by 111In-DTPA-octreotide and also reported greater detection of lesions by 64Cu-DOTATATE [36].
64Cu-DOTATATE is administered intravenously and has some adverse effects, including nausea, vomiting, and flushing [37]. It was developed by Radiomedix inc. and was approved by the FDA on 3 September 2020 [38].

3.3. 68Ga-PSMA-11

68Ga-PSMA-11 or (68Ga gozetotide) consists of the lipophilic acyclic chelator HBED-CC (N,N′-bis [2-hydroxy-5-(carboxyethyl)benzyl] ethylenediamineN,N′- diacetic acid) (blue), which is coordinated to the radionuclide 68Ga (purple) and a urea-based peptidomimetic HO-Glu-NH-CO-NH-Lys-OH (black). Both moieties are linked by a 6-aminohexanoic acid (green) through the side chain (ε-NH2) of the Lys residue (Figure 6) [39,40].
68Ga-PSMA-11 is indicated for PET imaging of prostate-specific membrane antigen (PSMA)-positive lesions in men with prostate cancer [41]. It is the first PET drug to trace and detect prostate carcinoma relapses and metastases with high contrast by targeting the extracellular domain of the PSMA. It is a transmembrane glycoprotein expressed in the prostate, kidneys, small intestines, salivary glands, and brain [39,40]. However, it is overexpressed 100- to 1000-fold in prostate tumors compared to other tissues [40,42], thereby offering a specific target for diagnostic and therapeutic purposes.
The active binding site of PSMA combines a hydrophilic motif that interacts with the urea-based inhibitor Glu-NH-CO-NH-Lys- (its role being to drive the radionuclide to the cancer cells with high specificity), and a lipophilic pocket. Thus, the lipophilic nature of the HBED-CC chelator has a positive impact on the binding properties. Moreover, the chelator has shown high efficiency to complex with Ga(III) [40]. Compared to the corresponding DOTA-conjugate of the same pharmacophore, the HBED-CC chelator-conjugate demonstrated improved PET imaging performance as it showed reduced unspecific binding and considerably higher specificity, fast blood, and organ clearances, and low accumulation in liver [40].
Other conjugates, i.e., labeled antibodies, can also be used for targeting the cytoplasmic PMSA, such as 111In-capromab pendetide (ProstaScint®), which is used for scintigraphy. However, accessibility to the cytoplasmic PMSA compared to the extracellular protein is lower, especially for large molecules. Furthermore, the long half-life of 111In and, consequently, the long length of exposure to radiation and the prolonged circulation of the antibody in the body, which causes high background noise, make PET imaging with 68Ga more favorable than the scintigraphic method [42].
68Ga-PSMA-11 is administered intravenously and has some adverse effects, including nausea, diarrhea, and dizziness [41]. It was developed by the University of California (Los Angeles) and was approved by the FDA on 1 December 2020 [43].

3.4. Belantamab Mafodotin-Blmf (BlenrepTM)

Belantamab mafodotin-blmf is the ninth ADC approved by the FDA. It is composed by afucosylated humanized immunoglobulin G1 monoclonal antibody (IgG1) (blue) and a peptide payload called monomethyl auristatin F (MMAF) (red) [44]. The two units are linked by the protease-resistant maleimidohexanoic linker (green) (Figure 7).
The afucosylated IgG1 antibody, which targets B-cell maturation antigen (BCMA) [45], is produced by recombinant DNA technology in a mammalian cell line (Chinese Hamster Ovary), whereas the payload-linker is prepared synthetically. Final attachment is by conjugation of the maleimide to the Cys residues on the antibody. To this end, a controlled reduction of various interchain disulfide bonds on the antibody must be achieved to prevent its denaturation. Usually, this step is done using the reducing agent tris-(2-carboxyethyl)-phosphine (TCEP), then the free thiol of such Cys residues react with maleimides under very mild conditions [46]. The conjugate obtained carries approximately four molecules of MMAF per antibody molecule.
MMAF is a synthetically modified analog of natural dolastatin 10 (Figure 8), which acts as an antimitotic agent, inhibiting cell division by blocking the polymerization of tubulin [45]. Due to the high cytotoxicity of both dolastatin 10 and its synthetic analogs, they cannot be administered directly but have to be delivered and deposited at the tumor site. The maleimidohexanoyl linker is considered non-cleavable because it is protease-resistant. Thus, catabolism of the mAb backbones is required to release the drug [47], in which a cysteine-adduct of the linker-MMAF derivative is released [44].
The ADCs approved in 2019 (Padcev and Polivy) used a monomethyl auristatin E (MMAE) analog as payload [8], which is more membrane-permeable than MMAF [48]. On the other hand, MMAF is more hydrophilic and this property decreases aggregation tendencies [48]. Most importantly, MMAF has lower bystander activities (drug release to neighboring antigen-negative cells or tissues) than MMAE [49]. Finally, MMAF shows improved pharmacokinetics and also a better therapeutic index [50] (Figure 8). Furthermore, the (Padcev and Polivy) were built using a cleavable linker. Of note, in contrast to ADCs with cleavable linkers, those with non-cleavable linkers have an extended plasma half-life as their bonds are not susceptible to lysosomal processes [51].
Belantamab mafodotin-blmf is prescribed for the treatment of adult patients with relapsed or refractory multiple myeloma who have received at least four prior therapies, including an anti-CD38 monoclonal antibody, a proteasome inhibitor, and an immunomodulatory agent [45]. Upon the binding of the mAb to the surface of the BCMA cell, belantamab mafodotin-blmf is internalized and MMAF is released via the proteolysis of the mAb component (as the linker is non-cleavable), ending with apoptosis [52].
Belantamab mafodotin-blmf is administered intravenously every three weeks [45,52]. It has various adverse effects: ≥20% are keratopathy (corneal epithelium change on eye exam), decreased visual acuity, nausea, blurred vision, pyrexia, infusion-related reactions, and fatigue; and ≥5% are decreased platelets, decreased lymphocytes, decreased hemoglobin, decreased neutrophils, increased creatinine, and increased gamma-glutamyl transferase.
Belantamab mafodotin-blmf was developed by GlaxoSmithKline and approved by the FDA on 5 August 2020 [53] and by the European Medicines Agency (EMA) on 25 August 2020 [52].

4. Conclusions

A total of 6 out of the 53 drugs approved by the FDA in 2020 belong to the TIDES category. The continued authorization of these chemical entities highlights their efficacy and safety profiles, and therefore the growing relevance of this family of drugs.
The peptide market has been strengthened over many years and this trend is expected to continue. During the preparation of this manuscript, it was disclosed that PlitidepsinTM (aplidin), a peptide of marine origin approved a few years ago in Australia for multiple myeloma [54], shows potent preclinical efficacy against SARS-CoV-2 [55] and is expected to enter clinical phase III very shortly. Futhermore, on 22 January 2021, voclosporin (LupkynisTM) was approved for the treatment of treat lupus nephritis [56]. Voclosporin is cyclosporine analogue.
Several newcomers to the oligonucleotide family, namely siRNAs and antisense oligonucleotides, are expected to join the pharmaceutical market arena shortly. Inclisiran, a siRNA for the treatment of hypercholesterolemia, was developed by Novartis under a license in collaboration with Alnylam Pharmaceuticals. It has been approved in Europe and is expected to receive authorization from the FDA soon [13,57,58]. Early members of the oligonucleotide drugs family demonstrated their efficacy in the treatment of some rare hereditary disorders. However, new promising candidates are now widening the applicability of this family for the treatment of more common diseases.
The applications of TIDES as drugs will undoubtedly continue to play an important role as drugs, as they are fuelled by the continuous research undertaken in both the academic and industrial setting.

Author Contributions

All authors participated in the search for information and in writing the manuscript, and approved the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded in part by the following: the National Research Foundation (NRF) and the University of KwaZulu-Natal (South Africa); MINECO, (RTI2018-093831-B-100), and the Generalitat de Catalunya (2017 SGR 1439) (Spain).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mullard, A. 2020 FDA drug approvals. Nat. Rev. Drug Discov. 2021. [Google Scholar] [CrossRef]
  2. De la Torre, B.G.; Albericio, F. The pharmaceutical industry in 2020. An analysis of FDA drug approvals from the perspective of molecules. Molecules 2021, 26, 627. [Google Scholar] [CrossRef]
  3. Defitelio Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/208114lbl.pdf (accessed on 28 January 2021).
  4. Dzierlega, K.; Yokota, T. Optimization of antisense-mediated exon skipping for duchenne muscular dystrophy. Gene Ther. 2020, 27, 407–416. [Google Scholar] [CrossRef] [PubMed]
  5. Dhillon, S. Viltolarsen: First approval. Drugs 2020, 80, 1027–1031. [Google Scholar] [CrossRef]
  6. Iftikhar, M.; Frey, J.; Shohan, M.J.; Malek, S.; Mousa, S.A. Current and emerging therapies for duchenne muscular dystrophy and spinal muscular atrophy. Pharmacol. Ther. 2020, 107719. [Google Scholar] [CrossRef]
  7. Komaki, H.; Takeshima, Y.; Matsumura, T.; Ozasa, S.; Funato, M.; Takeshita, E.; Iwata, Y.; Yajima, H.; Egawa, Y.; Toramoto, T.; et al. Viltolarsen in japanese duchenne muscular dystrophy patients: A phase 1/2 study. Ann. Clin. Transl. Neurol. 2020. [Google Scholar] [CrossRef]
  8. Al Shaer, D.; Al Musaimi, O.; Albericio, F.; de la Torre, B.G. 2019 FDA tides (peptides and oligonucleotides) harvest. Pharmaceuticals 2020, 13, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Viltepso Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212154Orig1s000lbl.pdf (accessed on 16 January 2021).
  10. Viltepso Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/212154Orig1s000ltr.pdf (accessed on 16 January 2021).
  11. Oxlumo Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/214103lbl.pdf (accessed on 16 January 2021).
  12. Scott, L.J. Givosiran: First approval. Drugs 2020, 80, 335–339. [Google Scholar] [CrossRef]
  13. Debacker, A.J.; Voutila, J.; Catley, M.; Blakey, D.; Habib, N. Delivery of oligonucleotides to the liver with galnac: From research to registered therapeutic drug. Mol. Ther. 2020, 28, 1759–1771. [Google Scholar] [CrossRef]
  14. Cochat, P.; Rumsby, G. Primary hyperoxaluria. N. Engl. J. Med. 2013, 369, 649–658. [Google Scholar] [CrossRef] [PubMed]
  15. Dindo, M.; Conter, C.; Oppici, E.; Ceccarelli, V.; Marinucci, L.; Cellini, B. Molecular basis of primary hyperoxaluria: Clues to innovative treatments. Urolithiasis 2019, 47, 67–78. [Google Scholar] [CrossRef]
  16. Liebow, A.; Li, X.; Racie, T.; Hettinger, J.; Bettencourt, B.R.; Najafian, N.; Haslett, P.; Fitzgerald, K.; Holmes, R.P.; Erbe, D.; et al. An investigational rnai therapeutic targeting glycolate oxidase reduces oxalate production in models of primary hyperoxaluria. J. Am. Soc. Nephrol. 2017, 28, 494–503. [Google Scholar] [CrossRef] [Green Version]
  17. Scott, L.J.; Keam, S.J. Lumasiran: First approval. Drugs 2021. [Google Scholar] [CrossRef] [PubMed]
  18. Oxlumo Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/214103Orig1s000ltr.pdf (accessed on 16 January 2021).
  19. Falls, B.A.; Zhang, Y. Insights into the allosteric mechanism of setmelanotide (rm-493) as a potent and first-in-class melanocortin-4 receptor (mc4r) agonist to treat rare genetic disorders of obesity through an in silico approach. ACS Chem. Neurosci. 2019, 10, 1055–1065. [Google Scholar] [CrossRef]
  20. Al Musaimi, O.; Al Shaer, D.; de la Torre, B.G.; Albericio, F. 2017 FDA peptide harvest. Pharmaceuticals 2018, 11, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Brayden, D.J.; Hill, T.A.; Fairlie, D.P.; Maher, S.; Mrsny, R.J. Systemic delivery of peptides by the oral route: Formulation and medicinal chemistry approaches. Adv. Drug. Deliv. Rev. 2020, 157, 2–36. [Google Scholar] [CrossRef] [PubMed]
  22. Clément, K.; Biebermann, H.; Farooqi, I.S.; Van der Ploeg, L.; Wolters, B.; Poitou, C.; Puder, L.; Fiedorek, F.; Gottesdiener, K.; Kleinau, G.; et al. Mc4r agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat. Med. 2018, 24, 551–555. [Google Scholar] [CrossRef] [PubMed]
  23. Imcivree Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/213793s000lbl.pdf (accessed on 16 January 2021).
  24. Ayers, K.L.; Glicksberg, B.S.; Garfield, A.S.; Longerich, S.; White, J.A.; Yang, P.; Du, L.; Chittenden, T.W.; Gulcher, J.R.; Roy, S.; et al. Melanocortin 4 receptor pathway dysfunction in obesity: Patient stratification aimed at mc4r agonist treatment. J. Clin. Endocrinol. Metab. 2018, 103, 2601–2612. [Google Scholar] [CrossRef]
  25. Clément, K.; Vaisse, C.; Lahlou, N.; Cabrol, S.; Pelloux, V.; Cassuto, D.; Gourmelen, M.; Dina, C.; Chambaz, J.; Lacorte, J.M.; et al. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 1998, 392, 398–401. [Google Scholar] [CrossRef]
  26. Collet, T.H.; Dubern, B.; Mokrosinski, J.; Connors, H.; Keogh, J.M.; Mendes de Oliveira, E.; Henning, E.; Poitou-Bernert, C.; Oppert, J.M.; Tounian, P.; et al. Evaluation of a melanocortin-4 receptor (mc4r) agonist (setmelanotide) in mc4r deficiency. Mol. Metab. 2017, 6, 1321–1329. [Google Scholar] [CrossRef]
  27. Haws, R.; Brady, S.; Davis, E.; Fletty, K.; Yuan, G.; Gordon, G.; Stewart, M.; Yanovski, J. Effect of setmelanotide, a melanocortin-4 receptor agonist, on obesity in bardet-biedl syndrome. Diabetes Obes. Metab. 2020, 22, 2133–2140. [Google Scholar] [CrossRef]
  28. Kühnen, P.; Clément, K.; Wiegand, S.; Blankenstein, O.; Gottesdiener, K.; Martini, L.L.; Mai, K.; Blume-Peytavi, U.; Grüters, A.; Krude, H. Proopiomelanocortin deficiency treated with a melanocortin-4 receptor agonist. N. Eng. J. Med. 2016, 375, 240–246. [Google Scholar] [CrossRef] [PubMed]
  29. Kievit, P.; Halem, H.; Marks, D.L.; Dong, J.Z.; Glavas, M.M.; Sinnayah, P.; Pranger, L.; Cowley, M.A.; Grove, K.L.; Culler, M.D. Chronic treatment with a melanocortin-4 receptor agonist causes weight loss, reduces insulin resistance, and improves cardiovascular function in diet-induced obese rhesus macaques. Diabetes 2013, 62, 490–497. [Google Scholar] [CrossRef] [Green Version]
  30. Imcivree Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/213793Orig1s000ltr.pdf (accessed on 16 January 2021).
  31. Gutfilen, B.; Souza, S.A.; Valentini, G. Copper-64: A real theranostic agent. Drug Des. Devel. Ther. 2018, 12, 3235–3245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Grassi, I.; Nanni, C.; Cicoria, G.; Blasi, C.; Bunkheila, F.; Lopci, E.; Colletti, P.M.; Rubello, D.; Fanti, S. Usefulness of 64cu-atsm in head and neck cancer: A preliminary prospective study. Clin. Nucl. Med. 2014, 39, e59–e63. [Google Scholar] [CrossRef]
  33. Handley, M.G.; Medina, R.A.; Mariotti, E.; Kenny, G.D.; Shaw, K.P.; Yan, R.; Eykyn, T.R.; Blower, P.J.; Southworth, R. Cardiac hypoxia imaging: Second-generation analogues of 64cu-atsm. J. Nucl. Med. 2014, 55, 488–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Jalilian, A.R.; Osso, J., Jr. The current status and future of theranostic copper-64 radiopharmaceuticals. Iran. J. Nucl. Med. 2017, 25, 1–10. Available online: https://irjnm.tums.ac.ir/article_23081.html (accessed on 1 February 2021).
  35. Johnbeck, C.B.; Knigge, U.; Loft, A.; Berthelsen, A.K.; Mortensen, J.; Oturai, P.; Langer, S.W.; Elema, D.R.; Kjaer, A. Head-to-head comparison of (64)cu-dotatate and (68)ga-dotatoc pet/ct: A prospective study of 59 patients with neuroendocrine tumors. J. Nucl. Med. 2017, 58, 451–457. [Google Scholar] [CrossRef] [Green Version]
  36. Pfeifer, A.; Knigge, U.; Mortensen, J.; Oturai, P.; Berthelsen, A.K.; Loft, A.; Binderup, T.; Rasmussen, P.; Elema, D.; Klausen, T.L.; et al. Clinical pet of neuroendocrine tumors using 64cu-dotatate: First-in-humans study. J. Nucl. Med. 2012, 53, 1207–1215. [Google Scholar] [CrossRef] [Green Version]
  37. Detectnet Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/213227s000lbl.pdf (accessed on 16 January 2021).
  38. Detectnet Aprroval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/213227Orig1s000ltr.pdf (accessed on 16 January 2021).
  39. Minamimoto, R.; Hancock, S.; Schneider, B.; Chin, F.T.; Jamali, M.; Loening, A.; Vasanawala, S.; Gambhir, S.S.; Iagaru, A. Pilot comparison of (6)(8)ga-rm2 pet and (6)(8)ga-psma-11 pet in patients with biochemically recurrent prostate cancer. J. Nucl Med. 2016, 57, 557–562. [Google Scholar] [CrossRef] [Green Version]
  40. Eder, M.; Schafer, M.; Bauder-Wust, U.; Hull, W.E.; Wangler, C.; Mier, W.; Haberkorn, U.; Eisenhut, M. 68ga-complex lipophilicity and the targeting property of a urea-based psma inhibitor for pet imaging. Bioconjug. Chem. 2012, 23, 688–697. [Google Scholar] [CrossRef]
  41. Gallium 68 psma-11 Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212642s000lbl.pdf (accessed on 16 January 2021).
  42. Afshar-Oromieh, A.; Malcher, A.; Eder, M.; Eisenhut, M.; Linhart, H.G.; Hadaschik, B.A.; Holland-Letz, T.; Giesel, F.L.; Kratochwil, C.; Haufe, S.; et al. Pet imaging with a [68ga]gallium-labelled psma ligand for the diagnosis of prostate cancer: Biodistribution in humans and first evaluation of tumour lesions. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 486–495. [Google Scholar] [CrossRef]
  43. Gallium 68 psma-11 Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/212642Orig1s000ltr.pdf (accessed on 16 January 2021).
  44. Bouchard, H.; Viskov, C.; Garcia-Echeverria, C. Antibody-drug conjugates-a new wave of cancer drugs. Bioorg. Med. Chem. Lett. 2014, 24, 5357–5363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Blenrep Drug Label. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761158s000lbl.pdf (accessed on 16 January 2021).
  46. Matsuda, Y.; Mendelsohn, B.A. An overview of process development for antibody-drug conjugates produced by chemical conjugation technology. Expert. Opin. Biol. Ther. 2020. [Google Scholar] [CrossRef] [PubMed]
  47. Han, T.H.; Zhao, B. Absorption, distribution, metabolism, and excretion considerations for the development of antibody-drug conjugates. Drug Metab. Dispos. 2014, 42, 1914–1920. [Google Scholar] [CrossRef]
  48. Park, M.H.; Lee, B.I.; Byeon, J.J.; Shin, S.H.; Choi, J.; Park, Y.; Shin, Y.G. Pharmacokinetic and metabolism studies of monomethyl auristatin f via liquid chromatography-quadrupole-time-of-flight mass spectrometry. Molecules 2019, 24, 2754. [Google Scholar] [CrossRef] [Green Version]
  49. Moquist, P.N.; Bovee, T.D.; Waight, A.B.; Mitchell, J.A.; Miyamoto, J.B.; Mason, M.L.; Emmerton, K.K.; Stevens, N.; Balasubramanian, C.; Simmons, J.K.; et al. Novel auristatins with high bystander and cytotoxic activities in drug-efflux positive tumor models. Mol. Cancer Ther. 2020. [Google Scholar] [CrossRef]
  50. Joubert, N.; Beck, A.; Dumontet, C.; Denevault-Sabourin, C. Antibody-drug conjugates: The last decade. Pharmaceuticals 2020, 13, 245. [Google Scholar] [CrossRef] [PubMed]
  51. Jain, N.; Smith, S.W.; Ghone, S.; Tomczuk, B. Current adc linker chemistry. Pharm. Res. 2015, 32, 3526–3540. [Google Scholar] [CrossRef] [Green Version]
  52. Tzogani, K.; Penttilä, K.; Lähteenvuo, J.; Lapveteläinen, T.; Lopez Anglada, L.; Prieto, C.; Garcia-Ochoa, B.; Enzmann, H.; Gisselbrecht, C.; Delgado, J.; et al. Ema review of belantamab mafodotin (blenrep) for the treatment of adult patients with relapsed/refractory multiple myeloma. Oncologist 2020. [Google Scholar] [CrossRef]
  53. Blenrep Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/761158Orig1s000ltr.pdf (accessed on 16 January 2021).
  54. El Bairi, K.; Trapani, D.; Petrillo, A.; Le Page, C.; Zbakh, H.; Daniele, B.; Belbaraka, R.; Curigliano, G.; Afqir, S. Repurposing anticancer drugs for the management of covid-19. Eur. J. Cancer 2020, 141, 40–61. [Google Scholar] [CrossRef] [PubMed]
  55. White, K.M.; Rosales, R.; Yildiz, S.; Kehrer, T.; Miorin, L.; Moreno, E.; Jangra, S.; Uccellini, M.B.; Rathnasinghe, R.; Coughlan, L.; et al. Plitidepsin has potent preclinical efficacy against sars-cov-2 by targeting the host protein eef1a. Science 2021. [Google Scholar] [CrossRef]
  56. Lupkynis Approval Letter. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2021/213716Orig1s000ltr.pdf (accessed on 1 February 2021).
  57. Novartis Successfully Completes Acquisition of the Medicines Company, Adding a Potentially First-in-Class, Investigational Cholesterol-Lowering Therapy Inclisiran. Available online: https://www.novartis.com/news/media-releases/novartis-successfully-completes-acquisition-medicines-company-adding-potentially-first-class-investigational-cholesterol-lowering-therapy-inclisiran (accessed on 16 January 2021).
  58. New Novartis Data for Inclisiran Shows Effective and Sustained ldl-c Reduction at 17 Regardless of Age or Gender. Available online: https://www.dicardiology.com/content/new-novartis-data-inclisiran-shows-effective-and-sustained-ldl-c-reduction-17-regardless-age (accessed on 16 January 2021).
Figure 1. A total of 228 new drugs were approved by the Food and Drug Administration (FDA) from 2016 to 2020 [2]. mAbs, monoclonal antibodies; ADCs, antibody drug conjugates; Oligos, oligonucleotides.
Figure 1. A total of 228 new drugs were approved by the Food and Drug Administration (FDA) from 2016 to 2020 [2]. mAbs, monoclonal antibodies; ADCs, antibody drug conjugates; Oligos, oligonucleotides.
Pharmaceuticals 14 00145 g001
Figure 2. Structure of viltolarsen (ViltepsoTM).
Figure 2. Structure of viltolarsen (ViltepsoTM).
Pharmaceuticals 14 00145 g002
Figure 3. Chemical structure of lumasiran (OxlumoTM).
Figure 3. Chemical structure of lumasiran (OxlumoTM).
Pharmaceuticals 14 00145 g003
Figure 4. Chemical structure of setmelanotide (ImcivreeTM).
Figure 4. Chemical structure of setmelanotide (ImcivreeTM).
Pharmaceuticals 14 00145 g004
Figure 5. Chemical structure of 64Cu -DOTATATE (DetectnetTM).
Figure 5. Chemical structure of 64Cu -DOTATATE (DetectnetTM).
Pharmaceuticals 14 00145 g005
Figure 6. Chemical structure of 68Ga-PMSA-11.
Figure 6. Chemical structure of 68Ga-PMSA-11.
Pharmaceuticals 14 00145 g006
Figure 7. Chemical structure of belantamab mafodotin-blmf (BlenrepTM).
Figure 7. Chemical structure of belantamab mafodotin-blmf (BlenrepTM).
Pharmaceuticals 14 00145 g007
Figure 8. Chemical structure of: (A) natural dolastatin 10; (B) monomethyl auristatin F (MMAF); and (C) monomethyl auristatin E (MMAE). Differences from the natural molecule are shown in red [44].
Figure 8. Chemical structure of: (A) natural dolastatin 10; (B) monomethyl auristatin F (MMAF); and (C) monomethyl auristatin E (MMAE). Differences from the natural molecule are shown in red [44].
Pharmaceuticals 14 00145 g008
Table 1. Summary of the 2020 FDA peptides and oligonucleotides (TIDES) harvest.
Table 1. Summary of the 2020 FDA peptides and oligonucleotides (TIDES) harvest.
#Active Ingredient (Trade Name)TypeIndicationTargetRoute
1Viltolarsen (ViltepsoTM)Antisense
oligonucleotide
Duchenne’s muscular dystrophy (DMD)DMD Exon 53Intravenous
2Lumasiran
(OxlumoTM)
Antisense
oligonucleotide
Primary
hyperoxaluria type 1 (PH1)
Hydroxyacid oxidase (glycolate oxidase) 1 (HOA1) mRNASubcutaneous
3Setmelanotide (ImcivreeTM)PeptideChronic weight
management (obesity)
Melanocortin-4
receptor (MC4R)
Subcutaneous
464Cu-DOTATATE (DetectnetTM)PeptideScintigraphic imagingSomatostatin receptorIntravenous
568Ga-PSMA-11PeptideDiagnosis of recurrent prostate carcinomaProstate-specific
membrane antigen (PSMA)
Intravenous
6Belantamab mafodotin-blmf (BlenrepTM)ADC a with peptide payload Relapsed or refractory multiple myelomaB-cell maturation
antigen (BCMA)
Intravenous
a ADC, antibody drug conjugate.
Table 2. FDA-approved oligonucleotides from 1998–2020.
Table 2. FDA-approved oligonucleotides from 1998–2020.
#Active Ingredient
(Trade Name)
CompanyStructureIndicationTargetFDA Approval
1Fomivirsen
(VitraveneTM)
Ionis Pharma
(Carlsbad, CA, USA)
Novartis
(Basel, Switzerland)
Antisense PS-ON aCytomegalovirus retinitisCMV UL123August 1998
2Pegaptanib
(MacugenTM)
NeXstar Pharma
(Boulder, CO, USA)
Eyetech Pharma
(Knolls, NJ, USA)
Polynucleotide aptamerNeovascular age-related macular degenerationVEGF-165December 2004
3Mipomersen
(KynamroTM)
Ionis Pharma
(Carlsbad, CA, USA)
Genzyme
(Cambridge, MA, USA)
Kastle Tx
(Housten, TX, USA)
Antisense PS-ON aHomozygous familial hypercholesterolaemiaAPOBJanuary 2013
4Defibrotide
(DefitelioTM)
Jazz Pharma
(Dublin, Ireland)
Mixed single strands of DNAHepatic veno-occlusive diseaseNA bMarch 2016
5Eteplirsen
(Exondys 51TM)
Sarepta Tx
(De Berry, TX, USA)
Antisense PS-ON a/(PMO c)Duchenne muscular dystrophyDMD exon 51September 2016
6Nusinersen
(SpinrazaTM)
Ionis Pharma
(Carlsbad, CA, USA)
Biogen
(Cambridge, MA, USA)
Antisense PS-ON a/(PMO c)Spinal muscular atrophySMN2 exon 7December 2016
7Patisiran
(OnpattroTM)
Alnylam Pharma
(Cambridge, MA, USA)
siRNA dHereditary transthyretin amyloidosis polyneuropathyTTRAugust 2018
8Inotersen
(TegsediTM)
Ionis Pharma
(Carlsbad, CA, USA)
Akcea Pharma
(Boston, MA, USA)
Antisense PS-ON aHereditary transthyretin; Amyloidosis polyneuropathyTTROctober 2018
9Givosiran
(GivlaariTM)
Alnylam Pharma
(Cambridge, MA, USA)
siRNA dAcute hepatic porphyriaAminolevulinate synthase 1 (ALAS1) mRNA eNovember 2019
(Enhanced)
10Golodirsen
(Vyondys 53TM)
Sarepta Tx
(De Berry, TX, USA)
Antisense PMO cDuchenne muscular dystrophyDMD exon 53December 2019
11Viltolarsen (ViltepsoTM)Nippon Shinyaku
(Kisshoin, Minami-ku Kyoto)
with (NCNP) f
(Kodaira, Tokyo)
Antisense PMO cDuchenne muscular dystrophyDMD exon 53August 2020
12Lumasiran
(OxlumoTM)
Alnylam Pharma
(Cambridge, MA, USA)
siRNA dPrimary hyperoxaluria type 1 (PH1)HOA1 mRNA eNovember 2020
a PS-ON: phosphorothioate oligonucleotide; b NA: not applicable, the mechanism of action is not well understood [3]; c PMO: phosphorodiamidate morpholino oligonucleotide; d siRNA: small interfering RNA; e mRNA: messenger RNA; f National Center of Neurology and Psychiatry.
Table 3. FDA-approved peptides between 2016 and 2020.
Table 3. FDA-approved peptides between 2016 and 2020.
#Active Ingredient (Trade Name)CompanyStructureIndicationTargetFDA
Approval
Free Peptides
1Lixisenatide
(AdlyxinTM)
Sanofi-Aventis
(Paris, France)
44 AAs Diabetes type (II)Glucagon-like peptide 1 receptorJuly 2016
2Plecanatide
(TrulanceTM)
Synergy Pharmaceuticals
(New York, NY, USA)
16 AAs 2 disulfidesChronic idiopathic constipation Guanylate cyclase-C January 2017
3Etelcalcetide
(ParsabivTM)
KAI Pharmaceuticals
(South of San Francisco, CA, USA)
Amgen
(Thousand Oaks, CA, USA)
7 AAs (all D) 1 disulfide intermolecular L-CysSecondary hyperparathyroidism in adult chronic kidney disease Calcium-sensing
receptor
February 2017
4Abaloparatide
(TymloTM)
Radius Health
(Boston, MA, USA)
34 AAsAnabolic agentParathyroid hormone 1 receptorApril 2017
5Semaglutide
(OzempicTM)
Novo Nordisk
(Måløv, Denmark)
31 AAs branched PEG-fatty acid Diabetes type (II)Glucagon-like peptide 1 receptorDecember 2017
6Macimorelin
(MacrilenTM)
Aeterna Zentaris
(Frankfurt, Germany)
3 residues pseudopeptideDiagnosis of adult growth hormone deficiencyGrowth hormone
secretagogue receptor type 1
December 2017
7Angiotensin II
(GiaprezaTM)
La Jolla Pharmaceutical
(San Diego, CA, USA)
8 AAsSeptic shock, diabetes mellitus, and acute renal failureType-1 angiotensin II receptorDecember 2017
8Afamelanotide
(ScenesseTM)
University of Arizona
(Tucson, Arizona, USA)
Clinuvel Inc.
(Menlo Park, CA, USA)
13 AAsErythropoietic protoporphyria Melanocortin 1 receptor October 2019
9Bremelanotide
(VyleesiTM)
Palatin Technology
(East Windsor, NJ, USA)
AMAG Pharmaceuticals
(Waltham, MA, USA)
7AAs cyclic sidechain to tail Hypoactive sexual desire disorderMelanocortin receptorsJune 2019
10Setmelanotide
(ImcivreeTM)
Rhythm Pharmaceuticals
(Boston, MA, USA)
8AAs Cyclic disulfide ObesityMelanocortin-4 receptor November 2020
Peptide-Chelator-radionuclide conjugates
11[177Lu]-DOTA-TATE (LutatheraTM)Advanced Accelerator Applications
(Millburn, NJ, USA)
7AAs Cyclic disulfide Gastroenteropancreatic neuroendocrine tumors Somatostatin receptorJanuary 2018
12[68Ga]-DOTATOCUniversity of Iowa Health Care
(Iowa City, IA, USA)
A 7AAs Cyclic disulfide PET imagingSomatostatin receptorAugust 2019
13[64Cu]-DOTATATE (DetectnetTM)Radiomedix Inc.
(Housten, TX, USA)
A 7AAs Cyclic disulfidePET imagingSomatostatin receptorSeptember 2020
14[68Ga]-PSMA-11University of California
(Oakland, CA, USA)
Peptidomemitic 2AAs urea linkedDiagnosis of recurrent prostate carcinoma by PETProstate-specific membrane antigenDecember 2020
Peptides in ADC’s
15Enfortumab vedotin-ejfv
(PadcevTM)
Astellas Pharma
(Northbrook, IL, USA)
5 residues with γ-AA Urothelial cancersNectin-4 receptorDecember 2019
16Polatuzumab vedotin-piiq
(PolivyTM)
Roche
(South of San Francisco, CA, USA)
5 residues with γ-AARefractory diffuse large B-cell lymphomaCD79b receptor expressed in mature BcellsJune 2019
17Belantamab mafodotin-blmf
(BlenrepTM)
GlaxoSmithKline
(Brentford, UK)
5 residues with γ-AARelapsed or refractory multiple myelomaB-cell maturation
antigen
August 2020
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Al Musaimi, O.; Al Shaer, D.; Albericio, F.; de la Torre, B.G. 2020 FDA TIDES (Peptides and Oligonucleotides) Harvest. Pharmaceuticals 2021, 14, 145. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020145

AMA Style

Al Musaimi O, Al Shaer D, Albericio F, de la Torre BG. 2020 FDA TIDES (Peptides and Oligonucleotides) Harvest. Pharmaceuticals. 2021; 14(2):145. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020145

Chicago/Turabian Style

Al Musaimi, Othman, Danah Al Shaer, Fernando Albericio, and Beatriz G. de la Torre. 2021. "2020 FDA TIDES (Peptides and Oligonucleotides) Harvest" Pharmaceuticals 14, no. 2: 145. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020145

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop