Next Article in Journal
Unveiling the Venom Composition of the Colombian Coral Snakes Micrurus helleri, M. medemi, and M. sangilensis
Previous Article in Journal
Oxidative Stress and DNA Damage in Pagrus major by the Dinoflagellate Karenia mikimotoi
Previous Article in Special Issue
Increasing the Passive Range of Joint Motion in Stroke Patients Using Botulinum Toxin: The Role of Pain Relief
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Botulinum Neurotoxins: From Toxin to Medicine

Spasticity and Movement Disorder-ReSTaRt Service Unit, Physical Medicine and Rehabilitation Department, Policlinico of Foggia, University of Foggia, 71122 Foggia, Italy
Toxins 2023, 15(10), 621; https://doi.org/10.3390/toxins15100621
Submission received: 10 October 2023 / Accepted: 17 October 2023 / Published: 20 October 2023
(This article belongs to the Special Issue Botulinum Neurotoxins: From Toxin to Medicine)
Ancient scientific manuscripts indicate that Dr. J. Kerner was the first, between 1817 and 1822, to suggest the possible therapeutic use of botulinum toxin, which he called “sausage poison”, describing it in the final chapter of his 1822 monograph; based on his previous experiences, he concluded that the toxin, applied in minimal doses, would lower or block the hyperactivity and hyperexcitability of the overactive nervous system.
Therefore, the name botulinum is strictly related to the word botuls (from the Latin word botulus, meaning “sausage”) named in this way by the German physician Muller in 1870, whereas at the end of 1800, E.P. Van Ermengem identified a bacteria and its toxin [1,2].
It is fascinating to consider that after almost 100 years, the botulinum toxin (BoNT) has finally become an important drug for many human diseases.
It is known that BoNT is a protein consisting of a heavy chain and a light chain linked via a disulfide bond. The activity of BoNTs involves a multistep process of heavy-chain-mediated cell binding to neuronal cells, endocytosis, and translocation of the light chain into the cytosol, followed by cleavage by the light chain of soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins. The cleavage of SNARE proteins by the zinc endopeptidase action of the light chain results in the disrupted fusion of synaptic vesicles containing neurotransmitters with the plasma membrane [3].
Botulinum neurotoxin is similar to a chemical scalpel that reduces the activity of cholinergic junctions inducing, for example, reversible muscle denervation.
Its possibility to have this effect is highly appreciated in the medical field, especially for spasticity, strabismus, blepharospasm, cervical dystonia, axillary hyper-hidrosis, glabellar lines, migraine, and neurogenic bladder [4].
Many serotypes produced by clostridium botulinum have been identified (A,B,C, D,E,F, and G), many of them with different subtypes, target proteins, and cleavage sites, but only BoNT/A and BoNT/B are used as medical therapies, showing safety and efficacy.
Therefore, “From toxin to medicine” is the title of this Special Issue of Toxins as many years have passed, and the efficacy of this toxin for therapeutic interventions has been fully described in several published scientific studies.
It is very important that this Special Issue includes the study of Shin-Ichiro Miyashita et al, as they investigated the efficacy of BoNT/CD, a mosaic toxin of BoNT/C and BoNT/D, assessing its potential as a therapeutic alternative to BoNT/A [5].
The authors showed, in a cultured neuron assay, that BoNT/CD cleaved syntaxin and SNAP-25 more than BoNT/C and BoNT/A, and after mouse digit abduction muscle injection, BoNT/CD induced dose-dependent muscle paralysis lasting ~21 days whereas BoNT/A-induced paralysis lasting ~30 days. BoNT/C failed to induce local paralysis without systemic toxicity.
The authors concluded that BoNT/CD may be a potential alternative for patients who do not respond to existing BoNT-based therapeutics.
Until now, in the clinical setting, commercial BoNT-A products have been formulated as a powder for reconstitution before injection with saline solution.
However, new products in a ready-to-use liquid formulation are appearing on the market. Elena Fonfria et al., described, in an interesting study, the suitability of a potency method that uses the BoCell® assay as a replacement for the LD50 assay to test aboBoNT-A in both powder and liquid formulations.
Following the assay validation and comparability studies presented here, the BoCell® assay has received approval from regulatory authorities in the European Union (both powder and liquid formulations) and in the USA (powder formulation) for establishing the potency, batch release, and stability purposes of aboBoNT-A, whereas submissions and approvals in other geographic areas are ongoing [6].
As the title of this Special Issue is “from toxin to medicine”, this implies that a linear progression has occurred; therefore, following the studies cited before, focused on biological effects and methods to demonstrate them, clinical studies produced by clinicians with great expertise on spasticity, movement disorders, and pain have been collected.
Bensmail and colleagues described the real-world use of BoNT-A for spasticity and neurogenic detrusor overactivity treatment on 105,206 multiple sclerosis subjects identified using the electronic health records of those treated with BoNT-A injection within striated muscle for spasticity and/or within the detrusor smooth muscle for neurogenic detrusor overactivity. The authors studied different numbers of subjects who received BoNT-A injection, ranging from >1 to >3 treatment, and reported several therapeutic results depending on the number and proportion of patients who received at least one BoNT-A injection, the number and proportion of patients who received at least three BoNT-A injections, and the time interval between two BoNT-A injections administered; moreover, they examined the rate of intradetrusor BoNT-A treatment, the mean number of intradetrusor BoNT-A injections per patient, and the time interval between two intradetrusor BoNT-A injections.
This study reinforces the importance of repeated BoNT-A injections to maintain a satisfactory therapeutic outcome against spasticity and neurogenic detrusor overactivity in subjects with multiple sclerosis [7].
One of the first indications BoNT therapy was approved by the FDA for was cervical dystonia, and currently BoNT is recommended as the first-line treatment for cervical dystonia (CD), with statistically significant improvements seen in 70–90% of patients in clinical studies [8]. Accurate diagnosis, appropriate pattern classification, physician experience, and correct BoNT-A dose and precision of injections are the most important factors contributing to the successful treatment of cervical dystonia.
The ultrasound guide for injection has been shown to be quick and painless, and it provides the real-time visualization of muscles and adjacent anatomical structures in different studies, especially for spasticity treatment, compared with other injection techniques [9].
In this Special Issue, Tyślerowicz and colleagues evaluated the efficacy of ultrasound-guided BoNT-A injections in comparison with anatomic landmarks in patients with cervical dystonia, using the Toronto Western Spasmodic Torticollis Rating Scale, Tsui modified scale, Craniocervical Dystonia Questionnaire, and Clinical Global Impression—Improvement scale. The authors concluded that ultrasound guidance might be helpful in improving the results of BoNT-A injections in cervical dystonia, reducing associated pain and disability [10].
Pain is the most common non-motor symptom of cervical dystonia, affecting 55–90% of patients with CD, and is rated as moderate or severe by 71% of patients; it is often the main reason patients with CD seek treatment and is a major contributor to disability and impaired QoL [11].
The efficacy of BoNT-A in relieving pain in patients with CD has been confirmed in analyses of controlled clinical trial data and in large-scale observational studies/registries of CD patients [12,13,14].
BoNT-A has been found to inhibit bradykinins, serotonin, potassium, prostaglandin E2, substance P, and the neuropeptide CGRP, reducing the sensitization of muscle nociceptors [15]. Moreover, BoNT injections have been used to treat muscle pain evoked by chemical C and A fiber stimulation because of the local production of lactate and acid PH induced by prolonged muscle activity and long-term contracture.
The published paper into this Special Issue described a pooling data from four studies in cervical dystonia adults treated with BoNT-A (incobotulinumtoxinA) for pain reduction [16].
Pain is also a great problem for spastic limb rehabilitation in subjects with upper motor neuron syndrome. The same origin and mechanisms seen for CD have been postulated for pain due to stroke or traumatic brain injury, so BoNT-A can be used for reducing pain and spasticity. The possibilities of increasing the passive range of motion (p-ROM) of the upper spastic limb, reducing spasticity, and improving posture and pain during p-ROM assessment, measured using the Numeric Rating Scale (NRS), have been hypothesized by Trompetto et al., in a retrospective study in 70 post-stroke patients treated with BoNT-A for upper limb hypertonia [17].
At the end of the study, the authors postulated that pain reduction after BoNT-A treatment can be induced via the muscle relaxant effect acting on spastic dystonia and/or via a specific action along the nociceptive pathway acting on the sensory neurons, thus permitting a passive range of motion increase in stroke survivors.
Finally, we know the reasons why many patients with spasticity want to be treated with BoNT-A; however, we do not know why many subjects have discontinued treatment. Cinone et al. evaluated the reasons for and determinants of BoNT-A discontinuation in patients with stroke, multiple sclerosis, spinal cord injury, and traumatic brain injury by conducting a retrospective study of 56 discontinuer patients treated with botulinum toxin for up to ten years.
The main reasons for discontinuation were logistic problems (due to distance or the absence of an adequate caregiver) and surgical interventions for spasticity, including intrathecal baclofen.
The authors concluded that it is crucial to identify the possible predictors of discontinuation to improve the efficacy of multidisciplinary management while also confirming the crucial roles of rehabilitation and caregivers in achieving better long-term outcomes [18].

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Kerner, J. Das Fettgift oder die Fettsäure und Ihre Wirkungenauf den Thierischen Organismus, ein Beytrag zur Untersuchung des in Verdorbenen Wursten Giftig Wirkenden Stoffes; Cotta: Stuttgart, Germany; Tubingen, Germany, 1822. [Google Scholar]
  2. Erbguth, F.J.; Naumann, M. Historical aspects of botulinum toxin: Justinus Kerner (1786–1862) and the “sausage poison”. Neurology 1999, 53, 1850–1853. [Google Scholar] [CrossRef]
  3. Humeau, Y.; Doussau, F.; Grant, N.J.; Poulain, B. How botulinum and tetanus neurotoxins block neurotransmitter release. Biochimie 2000, 82, 427–446. [Google Scholar] [CrossRef] [PubMed]
  4. Simpson, D.M.; Gracies, J.-M.; Graham, H.K.; Miyasaki, J.M.; Naumann, M.; Russman, B.; Simpson, L.L.; So, Y. Assessment: Botulinum neurotoxin for the treatment of spasticity (an evidence based review): Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology 2008, 70, 1691–1698. [Google Scholar] [CrossRef] [PubMed]
  5. Miyashita, S.I.; Karatsu, S.; Fujiishi, M.; Huang, I.H.; Nagashima, Y.; Morobishi, T.; Hosoya, K.; Hata, T.; Dong, M.; Sagane, Y. Characterization of Serotype CD Mosaic Botulinum Neurotoxin in Comparison with Serotype C and A. Toxins 2023, 15, 123. [Google Scholar] [CrossRef] [PubMed]
  6. Fonfria, E.; Marks, E.; Foulkes, L.M.; Schofield, R.; Higazi, D.; Coward, S.; Kippen, A. Replacement of the Mouse LD50 Assay for Determination of the Potency of AbobotulinumtoxinA with a Cell-Based Method in Both Powder and Liquid Formulations. Toxins 2023, 15, 314. [Google Scholar] [CrossRef] [PubMed]
  7. Bensmail, D.; Karam, P.; Forestier, A.; Loze, J.Y.; Lévy, J. Trends in Botulinum Toxin Use among Patients with Multiple Sclerosis: A Population-Based Study. Toxins 2023, 15, 280. [Google Scholar] [CrossRef] [PubMed]
  8. Jankovic, J.; Adler, C.H.; Charles, P.D.; Comella, C.; Stacy, M.; Schwartz, M.; Sutch, S.M.; Brin, M.F.; Papapetropoulos, S. Rationale and design of a prospective study: Cervical Dystonia Patient Registry for Observation of Ona Botulinumtoxin a Efficacy (CD PROBE). BMC Neurol. 2011, 11, 140. [Google Scholar] [CrossRef] [PubMed]
  9. Santamato, A.; Micello, M.F.; Panza, F.; Fortunato, F.; Baricich, A.; Cisari, C.; Pilotto, A.; Logroscino, G.; Fiore, P.; Ranieri, M. Can botulinum toxin type A injection technique influence the clinical outcome of patients with post-stroke upper limb spasticity? A randomized controlled trial comparing manual needle placement and ultrasound-guided injection techniques. J. Neurol. Sci. 2014, 347, 3943. [Google Scholar]
  10. Tyślerowicz, M.; Dulski, J.; Gawryluk, J.; Sławek, J. Does Ultrasound Guidance Improve the Effectiveness of Neurotoxin Injections in Patients with Cervical Dystonia? (A Prospective, Partially Blinded, Clinical Study). Toxins 2022, 14, 674. [Google Scholar] [PubMed]
  11. Charles, P.D.; Adler, C.H.; Stacy, M.; Comella, C.; Jankovic, J.; Manack Adams, A.; Schwartz, M.; Brin, M.F. Cervical dystonia and pain: Characteristics and treatment patterns from CD PROBE (Cervical Dystonia Patient Registry for Observation of OnabotulinumtoxinA Efficacy). J. Neurol. 2014, 261, 1309–1319. [Google Scholar] [CrossRef] [PubMed]
  12. Marciniec, M.; Szczepańska-Szerej, A.; Papuć, E.; Rejdak, K. Targeting pain in the long-term treatment of cervical dystonia with botulinum toxin A. Int. J. Neurosci. 2022, 132, 1026–1030. [Google Scholar] [CrossRef] [PubMed]
  13. Charles, P.D.; Manack Adams, A.; Davis, T.; Bradley, K.; Schwartz, M.; Brin, M.F.; Patel, A.T. Neck pain and cervical dystonia: Treatment outcomes from CD PROBE (Cervical Dystonia Patient Registry for Observation of OnabotulinumtoxinA Efficacy). Pain Pract. 2016, 16, 1073–1082. [Google Scholar] [CrossRef] [PubMed]
  14. Trosch, R.M.; Espay, A.J.; Truong, D.; Gil, R.; Singer, C.; LeWitt, P.A.; Lew, M.F.; Tagliati, M.; Adler, C.H.; Chen, J.J.; et al. Multicenter observational study of abobotulinumtoxinA neurotoxin in cervical dystonia: The ANCHOR-CD registry. J. Neurol. Sci. 2017, 376, 84–90. [Google Scholar] [CrossRef] [PubMed]
  15. Aoki, K.R. Review of a proposed mechanism for the antinociceptive action of botulinum toxin type A. Neurotoxicology 2005, 26, 785–793. [Google Scholar] [CrossRef] [PubMed]
  16. Albanese, A.; Wissel, J.; Jost, W.H.; Castagna, A.; Althaus, M.; Comes, G.; Scheschonka, A.; Vacchelli, M.; Jinnah, H.A. Pain Reduction in Cervical Dystonia Following Treatment with IncobotulinumtoxinA: A Pooled Analysis. Toxins 2023, 15, 333. [Google Scholar] [CrossRef] [PubMed]
  17. Trompetto, C.; Marinelli, L.; Mori, L.; Bragazzi, N.; Maggi, G.; Cotellessa, F.; Puce, L.; Vestito, L.; Molteni, F.; Gasperini, G.; et al. Increasing the Passive Range of Joint Motion in Stroke Patients Using Botulinum Toxin: The Role of Pain Relief. Toxins 2023, 15, 335. [Google Scholar] [CrossRef] [PubMed]
  18. Cinone, N.; Santoro, L.; Spina, S.; Facciorusso, S.; Battaglia, M.; Baricich, A.; Marcogiuseppe, P.; Santamato, A. Reasons and Determinants of BoNT-A Treatment Discontinuation in Patients Living with Spasticity: A 10-Year Retrospective Analysis. Toxins 2022, 14, 675. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Santamato, A. Botulinum Neurotoxins: From Toxin to Medicine. Toxins 2023, 15, 621. https://doi.org/10.3390/toxins15100621

AMA Style

Santamato A. Botulinum Neurotoxins: From Toxin to Medicine. Toxins. 2023; 15(10):621. https://doi.org/10.3390/toxins15100621

Chicago/Turabian Style

Santamato, Andrea. 2023. "Botulinum Neurotoxins: From Toxin to Medicine" Toxins 15, no. 10: 621. https://doi.org/10.3390/toxins15100621

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