NL2027321B1 - Treatment of hyperinflammatory syndrome - Google Patents

Treatment of hyperinflammatory syndrome Download PDF

Info

Publication number
NL2027321B1
NL2027321B1 NL2027321A NL2027321A NL2027321B1 NL 2027321 B1 NL2027321 B1 NL 2027321B1 NL 2027321 A NL2027321 A NL 2027321A NL 2027321 A NL2027321 A NL 2027321A NL 2027321 B1 NL2027321 B1 NL 2027321B1
Authority
NL
Netherlands
Prior art keywords
receptor antagonist
mammalian
use according
lidocaine
administration
Prior art date
Application number
NL2027321A
Other languages
Dutch (nl)
Inventor
Hasan Djohan
Original Assignee
Hasan Djohan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to NL2027321A priority Critical patent/NL2027321B1/en
Application filed by Hasan Djohan filed Critical Hasan Djohan
Priority to IL296719A priority patent/IL296719A/en
Priority to JP2022559877A priority patent/JP2023521623A/en
Priority to CA3178140A priority patent/CA3178140A1/en
Priority to EP21717576.9A priority patent/EP4125843A1/en
Priority to AU2021248263A priority patent/AU2021248263B2/en
Priority to US17/907,640 priority patent/US20230128348A1/en
Priority to BR112022019824A priority patent/BR112022019824A2/en
Priority to PCT/NL2021/050208 priority patent/WO2021201680A1/en
Priority to CN202180025176.0A priority patent/CN115605194A/en
Application granted granted Critical
Publication of NL2027321B1 publication Critical patent/NL2027321B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Transplantation (AREA)
  • Molecular Biology (AREA)
  • Pain & Pain Management (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Described is an antagonist of a mammalian P2X7 receptor for use in the treatment of a 5 hyperinflammatory syndrome in a mammalian patient, by primary lymph node targeted administration of the said P2X7 receptor antagonist in the said patient to a concentration in the said targeted lymph nodes that is above the maximal tolerable plasma level of the said antagonist in the said mammal.

Description

Treatment of hyperinflammatory syndrome The invention relates to an antagonist of a mammalian P2X; receptor for use in the treatment of a hyperinflammatory syndrome in a mammalian patient, by primary lymph node targeted administration of the said P2X; receptor antagonist in the said patient to a concentration in the said targeted lymph nodes that is above the maximal tolerable plasma level of the said antagonist in the said mammal. Background of the invention A hyperinflammatory syndrome, or hyperinflammation, is a known phenomenon in the medical art and is a symptom of a vast plurality of diseases resulting in dramatic if not lethal effects for the patient. The term ‘hyperinflammation’ as used herein is defined by the following 6 criteria (Webb et al., Lancet Rheumatol 2020, 2, (12) 754-763): (1) Fever, defined as a temperature of more than 38.0°C; (2) Macrophage activation, defined as a ferritin concentration of 700 ug/l or more; (3) Haematological dysfunction, defined as a neutrophil to lymphocyte ratio of 10 or more or both haemoglobin concentration of 9.2 g/dl or less and platelet count of 110 x 19° cells/L or less; (4) Coagulopathy, defined as a D-dimer concentration of 1.5 ug/ml or more; (5) Hepatic injury, defined as a lactate dehydrogenase concentration of 400 U/L or more, or an aspartate aminotransferase concentration of 100 U/L or more; (6) Cytokinaemia, defined as an interleukin-6 concentration of 15 pg/ml or more, or a triglyceride concentration of 150 mg/dl or more, or a CRP concentration of 15 mg/dl or more.
Dyspnoea and pneumonia are common and abundant symptoms coinciding with hyperinflammation, in particular as a result of airways infections. A major event for hyperinflammation to occur is the release of intracellular ATP. The intercellular signalling by nucleotides (ATP, ADP, UTP and UDP) and nucleoside (adenosine) is known in the art as purinergic signalling. Under normal resting conditions the extracellular levels of ATP are quite low at nanomolar concentrations (2-3 nM), whereas under specific conditions ATP release can rise by more than 1000-fold; such conditions may occur for the diseases as mentioned in the corresponding section ‘diseases involving hyperinflammation’ below, and include e.g. inflammation reactions, mechanical stress, surfactant release, membrane depolarisation, hypoxia. In purinergic signalling, a plurality of receptors are known, for the ligand adenosine, also known as P1 receptors, and for nucleotide ligands, known as P2 receptors. The required extracellular concentrations of the ligand to reach an effect half way between baseline and maximal effect (half maximal effective dose — ECs) for adenosine is in the nanomolar range, whereas for ATP, UTP or ADP these concentrations range from 0.01 to 10 pM. All these receptors, are known to be subject to desensitisation. Desensitisation of a receptor is defined as being unresponsive to activation by the ligand, resulting in zero transmembrane anion current. However, one of the P2 receptors, the P2X; receptor, is not pone to desensitisation, and the ECso for ATP to activate this receptor is much higher, namely at >1 mM. At such an ATP level, all other P1 and P2 purinergic receptors are fully desensitized.
By a disease as described above, such as a severe infection, massive extracellular ATP is released by the infected cells. This may be confined to the airway mucosa and the lung or may be extensive in multiple organs. The extracellular ATP has been observed to accumulate to 1.4 mM (Zhao et al., Front Immunol 2019, 10, 2524), resulting in the vigorous activation of the P2X; receptors causing hyperinflammation with massive pro-inflammatory immune response, massive pro-inflammatory and anti-inflammatory cytokine release and large pore formation with tissue cell destruction (Savio et al., Front Pharmacol 2018, 9, 52).
As a result of desensitisation of theP1 and P2 receptors, the physiological inflammatory response is deactivated (known as immune paralysis), rendering the patient susceptible to secondary infections.
Regulatory T-cells (Tregs) are key elements in the control of hyperinflammation, accelerating adenosine generation from extracellular ATP. Activation of P2X; receptors inhibits the suppressive potential and stability of Tregs.
The P2X; receptor plays an important role in many chronic and acute diseases. These diseases may be confined to 1 organ (Alzheimer's disease, multiple sclerosis, colitis) or may be diffusely disseminated as in bacterial sepsis or severe microbial infections, such as COVID-19, see also the below section ‘diseases involving hyperinflammation’.
The current treatments of hyperinflammation are actually anti-inflammatory treatments. The drugs literally block the activation of the immune response by the inhibition of one or more pro- inflammatory pathways. These treatments undermine the physiological function of the pro- inflammatory immune response, namely to recognise an “attack” by the invading microorganisms (“alarm phase”) followed by the activation of the first line of defence (the innate immune system) and when required the activation of the adaptive immune system to specifically disarm the invader. Examples of such drug that hamper the inflammatory response of the patients are dexamethasone, baricitinib and anakinra.
Treatment of hyperinflammation has hitherto therefore been cumbersome. The present invention now provides a method for treatment of patients suffering from hyperinflammation without hampering the inflammatory response of the patients or at least to a much lesser extent.
It has been suggested in the art that the P2X; receptor could be a good candidate to target when treating hyperinflammation and concomitant dyspnoea and pneumonia. A P2X; receptor antagonist would block the vigorous activation of the P2X; receptors. Because a large proportion of the ATP release to the extracellular space is mediated by the P2X; receptors, antagonism thereof would result in the decrease of the extracellular ATP concentrations. This can potentially abrogate hyperinflammation and the concomitant immune paralysis. In addition, inhibition of P2X7 receptors has been described to promote the cell-autonomous conversion of CD4+ T cells into Tregs after stimulation of their T-cell receptors (Schenk et al., Sci Signal 2011, 4 (162) ra12). Amelioration of hyperinflammation by P2X; receptor inhibition appears to be based on the increased activation and clonal expansion of the anti-inflammatory Tregs population.
Many P2X; receptor antagonists have been identified thus far (North and Jarvis, Mol Phar, 2013 (83) 759-769; Sluyter, Adv Exp Med Biol Prot Rev 2017 (19) 17 - 53). In order to achieve an effect, these antagonists have been administered systemically, in order to be transported by the blood to the envisaged site of action.
For example, CE-224,535 500 (Pfizer), AZD9056 (Astra-Zeneca) and JNJ54175448 (Johnson & Johnson) have been administered orally, however without great success. The anaesthetic lidocaine has been reported to be a P2X; receptor antagonist (Okura et al., Anesth Analg 2015, 120 (3), 597 - 605). Additional P2X; receptor antagonists are listed in the below section 'P2X7 receptor antagonists’.
Although a P2X; receptor antagonist can abrogate hyperinflammation and restore the capacity of the immune system to combat secondary co-infections and improve the clinical condition in critically ill patients suffering from a severe airway infection, the problem with these compounds lie in the fact that in order to have an effect, the antagonist should bind the P2X; receptor to such an extent that the hyperinflammation and preferably the concomitant effects of e.g. dyspnoea are counteracted effectively. Such an effect may already be observed at a concentration of the receptor antagonist to inhibit the receptor for 10% (the so-called 1C1 value). The preferred inhibition is a 50% receptor inhibition, i.e. at the 1Cs value. However, for P2X; receptor antagonists, such a concentration is above the maximal tolerable plasma level of the said antagonist, i.e. resulting in undesired side effects such as anxiety, dizziness or even decreased spinal reflexes or worse. For example, the maximal tolerable plasma levels for lidocaine for humans are about 4.7 ug/ml, see table 1: Table 1: maximal tolerable plasma level for lidocaine in humans
EEE vomiting, twitching and tremors, seizures with reduced 0.080 18.74 consciousness, respiratory depression, coma, etc.)
For each P2X; receptor antagonist, the skilled person will be aware as how to determine the maximal tolerable plasma level.
However, P2X; receptor antagonists have not effectively been used for treatment of hyperinflammation, as in order to be effective, the systemic dose would exceed the maximal tolerable plasma level by far.
The present invention now provides P2X; receptor antagonists for use in the treatment of a hyperinflammatory syndrome in a mammalian patient, by primary lymph node targeted administration of the said P2X; receptor antagonist in the said patient to a concentration in the said targeted lymph nodes that is above the maximal tolerable plasma level of the said antagonist in the said mammal. By primary lymph node targeting, the envisaged IC, value can be obtained in the lymph nodes, while avoiding exceeding the maximal tolerable plasma level. The inventors have found that establishing the envisaged IC, value in lymph nodes results in effective treatment of hyperinflammation, and significantly relieves dyspnoea in patients suffering from severe airway infections, and other symptoms of hyperinflammation. Targeting lymph nodes was envisaged as it was contemplated that the lymphatic system is populated exclusively by trafficking immune cells, i.e. naive T-cells, activated T-cells, B-cells, dendritic cells, monocytes, macrophages, neutrophils, mast cells, eosinophils, basophils and other immunologically relevant cells. It was found that by selective inhibition of the P2X; receptors of the immune cells of the lymphatic system by a P2X7 receptor antagonist, clonal expansion of Tregs is induced. Subsequently, these Tregs migrate throughout the body exerting anti-inflammatory activity reducing systemic and (distant) local hyperinflammation.
The term ‘primary lymph node targeted’ refers to an administration or delivery route wherein the majority of the receptor antagonist is delivered directly from the administration site to the lymph node, while the effective amount of the said receptor antagonist in the plasma is at least 5 times, preferably at least 10 times or at least 15 times less than in the lymph node.
The antagonist is administered to a concentration in the targeted lymph nodes that corresponds to the IC, for the said receptor, the said IC, being above the maximal tolerable plasma level of the said antagonist in the said mammal, wherein x = 10, preferably = 20, more preferably = 30, even more preferably = 40 and most preferably about 50. At ICs, 10% receptor inhibition is observed, at IC2, 20% receptor inhibition is observed, and so on. The higher x, the more receptor inhibition, the more effective the hyperinflammation is treated. It is clear to the skilled person that for a receptor antagonist that binds stronger to the receptor, the IC value will be lower than for a receptor antagonist that binds weaker to the receptor. The stronger the antagonist binds, the less amount of the said antagonist is need to have the same effect as compared to a weaker binding antagonist. The IC-value is preferably determined as described in Okura, supra.
The skilled person will be aware of suitable delivery and administration routes for lymph node targeted administration. Preferred are topical and invasive administration. Invasive administration may not be suitable outside hospital settings.
Therefore, it is very attractive to administer the receptor antagonist by topical administration.
As a topical route, transmucosal and transdermal administration are preferred.
In such a case, the antagonist is preferably administered in a lipophilic form, as a hydrophilic form would tend to be preferentially absorbed in the blood, 5 resulting in undesired elevation of the plasma level of the receptor antagonist in the blood, and to less delivery in the lymph nodes.
To this end, the receptor antagonist is preferably in the form of the free base thereof.
In a very attractive embodiment, the lymph node targeted administration comprises transmucosal administration in a body cavity covered with mucosa, preferably the said mucosa are close to one or more lymph nodes in order to enable fast and direct delivery.
In particular, the oral cavity is suited for such administration.
However, both nasal and al administration is also possible.
With regard to nasal delivery, care has to be taken to preferably not inhale the receptor antagonist or to a minimal extent, as such inhalation may cause undesired elevation of the plasma level of the receptor antagonist.
When administered to the oral cavity, the administration is preferably buccal, sublingual, pharyngeal or a combination thereof.
The mucosa preferably have a low systemic permeability and are in close vicinity to lymph nodes.
Permeability of different mucosal tissues is e.g.described in Goyal et al., Nanomed Biotechnol 2018, 46 (sup2), 539-551 and Lesch, et al., J Dent Res 1989, 68(9), 1345-1349. It has been contemplated in the art that oral administration is inefficient route of drug delivery (Di Vergilio et al., Br J Pharmacol 2020). However, sublingual and buccal administration of the receptor antagonists, in particular lidocaine, has now been shown to be very effective without significantly elevating the antagonist level in the plasma.
The permeability of the skin and mucosa to water, drugs, etc. is reported to be dependent on the site of the administration.
For example, the permeability constant of the floor of the mouth (sublingual mucosa), lateral border of the tongue and buccal mucosa for tritium-labelled water is 22, 17 and 13 times as high as human skin, respectively.
Moreover, the capacity of the submucosal capillaries to absorb molecules is much higher than the subcutaneous capillaries.
Lidocaine hydrochloride is highly soluble in water (solubility of 680 mg/ml in water) and therefore will mainly be absorbed by the submucosal capillary.
In contrast, the high lipophilic lidocaine base (solubility of 4 mg/ml in water, 760 mg/ml in 95% ethanol and 790 mg/ml in chloroform is preferably absorbed by the local initial lymphatics in the submucosal tissue (Gröningsson, et al., In Analytical Profiles of Drug Substances, Florey, K., Ed.
Academic Press: 1985; Vol. 14, pp 207-243). In addition, the lymphatic drainage of the floor of the mouth is extensive, involving a large number of lymph nodes.
Sublingual and a buccal administration of lipophilic lidocaine base or of any other P2X; receptor antagonist is preferred.
With a high concentration in a relatively low total dose the ICso of the P2X; receptors in the draining lymph nodes can be achieved to control systemic hyperinflammation and avoid toxic plasma levels of lidocaine or any other P2X; receptor antagonist.
It is to be noted that sublingual and buccal administration of lipophilic lidocaine are different from oral administration of lidocaine.
Oral administration of lidocaine is aimed at the resorption of the drug in the gastrointestinal tract, i.e. to systemic administration.
In another embodiment, the administration is transdermal and in the form of a cream, ointment or lotion, patch or plaster and/or involves microneedles or a combination thereof.
For this type of administration, the receptor antagonist is preferably lipophilic for the same reason as described above.
Transdermal administration of P2X; receptor antagonist, in particular in lipophilic from, an optionally in combination with skin penetration enhancers, such as alpha- terpineol, ethanol, lipid based nanoformulation can provide for convenient application.
In another embodiment, the administration is invasive, in particular chosen from intradermal, subdermal or subcutaneous administration.
The dermal capillaries can transport substances from blood to tissue but the reabsorption of substances from tissue to blood is, if any, extremely low.
Apparently, specialised initial lymphatics harbouring one way valve leaflets capable of absorbing fluid and molecules from the interstitium are localised in the dermis.
The absorbed lymph fluid is then propelled forward in the lymphatic network by collecting lymphatic vessels harbouring a rhythmic contracting muscle layer.
This system brings fluids and particles into the lymph nodes where numerous immune processes take place.
The absorption of intradermal application into the lymph nodes appear to be 10 times slower than after deep subcutaneous application and leads to higher concentrations in the lymph nodes related to these lymphatic vessels.
Smaller particles migrate more rapidly towards the lymphatic vessels and lymphatic nodes than larger particles.
The route and rate of clearance after intradermal and subcutaneous administration in the back of the hand in humans resulted in clearance of the administered compounds after subcutaneous injection of 1 %/min. and after intradermal injection of 8 - 10 %/min.
The additional advantage is that the plasma concentrations of subcutaneously administered lidocaine are much lower than intravenously administered lidocaine.
Intravenous administration of 2 mg/kg lidocaine in cats is almost immediately followed by a peak plasma concentration of 3.6 Hg/mL (Thomasy et al., Am J Vet Res 2005, 66 (7), 1162-1166). In contrast, the achieved mean peak plasma concentrations after the subcutaneous administration of 30 mg/kg, 20 mg/kg and 10 mg/kg lidocaine are much lower: 1.69, 1.07 and 0.77 pg/mL, respectively (Hatef et al., Aesthet Surg J 209 (2), 122-128). The applied subcutaneous dose is 15, 10 and 5 times higher than the intravenous dose, respectively.
The difference in the plasma concentrations after intravenous and subcutaneous administration of lidocaine is caused by the fact that, in contrast to the intravenous administration, a large proportion of the subcutaneously administered lidocaine is drained into the lymphatic system.
This slows down the release of lidocaine to the venous blood.
Lymphatic absorption after intradermal administration is much higher than after deep subcutaneous administration.
As intradermal infusion with lidocaine is not an accepted administration route for lidocaine, subdermal administration of lidocaine is proposed using a catheter inserted just beneath the dermis, that will result in higher concentrations of lidocaine in the draining local lymph nodes than a deep subcutaneous or intravenous infusion.
For invasive administration according to the invention, the receptor antagonist is preferably hydrophilic in particular in the form of a water soluble pharmaceutically acceptable salt thereof, such as the chloride salt.
In another embodiment, the administration is intravenous, the antagonist being lipophilic and confined in a drug delivery system avoiding direct release in the blood, e.g. by using nano-sized drug delivery systems, liposomes or polymer micelles. Oral administration of a P2X; receptor antagonist is also possible using delivery systems for intestinal lymphatic drug transport such as chylomicrons, etc, where delivery to the plasma is avoided. Intravenous administration at a low dosage where the maximum tolerable plasma level is not exceeded results in, if any, a much less pronounced effect. For lidocaine, an intravenous administration of 0.6 mg/kg/hr can be applied.
In particular, the P2X; receptor activation is activated by extracellular ATP. However, P2X; receptors can also be activated by membrane stretching, proteins from apoptotic cells, LL-37, cathelicidin and antimicrobial peptides. It is to be noted that all forms of such receptor activations are antagonised by the P2X; receptor antagonist.
In an attractive embodiment, the administration is an immediate release dosage form or a sustained release dosage form.
The administration preferably comprises one or more bolus administrations or comprises continuous administration or a combination thereof. A bolus is to be understood as the administration of a single tablet, pouch, injection, aerosol etc., or of a plurality thereof in any combination when administered subsequently without significant time intervals therebetween. It is also attractive to administer in a continuous fashion, e.g. as an infusion, or as a combination between one or more bolus administrations and a continuous administration.
In a particular embodiment, a bolus dosage corresponds with at least 1,000 times the amount of the receptor antagonist, that is comprised in 1 ml plasma at the maximal tolerable plasma level of the said antagonist, preferably at least 5,000 times, more preferably at least 10,000 times. Is means that according to this embodiment, the bolus is defined by amount of the receptor antagonist that is present in 1 ml plasma at the maximum tolerable plasma level. For example, the maximal tolerable plasma level of lidocaine in humans is 4,7 ug/ml. this would mean that the bolus would be at least 1,000 times 4,7 kg, i.e. 4,7 mg.
The bolus is preferably administered 2 — 10 times daily.
The antagonist is preferably administered in a liquid medium comprising at least 1 w/v % of the receptor antagonist, preferably at least 5 w/v% and most preferably at least 10 w/v%. Such high concentrations of receptor antagonist, in particular of lidocaine have hitherto not been used in the art. Such concentrations, when used according to the art, i.e. directed to systemic delivery via the blood would lead to unacceptably high plasma levels of beyond the maximal tolerable plasma level of the said antagonist. For example, in order to treat hyperinflammation and concomitant dyspnoea in a patient suffering from severe airways infection e.g. by infection by SARS-CoV-2.
When it comes to invasive administration, the lymph node targeted administration is preferably by continuous intradermal, subdermal or subcutaneous infusion.
In particular patients that are intubated for ventilation and/or kept in coma may need such administration route.
For administration by a continuous infusion, the dosage preferably corresponds with at least times the IC+ value per kg body weight per hour, more preferably at least 10 times the Cz value per kg body weight per hour, even more preferably at least 10 times the ICs; value per kg body weight per hour, still even more preferably at least 10 times the IC4 value per kg body weight per hour, at most preferably least 10 times the IC:o value per kg body weight per hour, at least 10 times the IC+ value per kg body weight per hour, at least 10 times the IC49 value per kg 10 body weight per hour.
More preferably the dosage is at least 15 times the ICso value per kg body weight per hour.
For lidocaine, the latter value would correspond with about 1 mg/kg/hr.
The ICso value for lidocaine is 66 pg/ml (0.066 x 15 = 0.99). According to a very attractive embodiment, the treatment involves a hyperinflammatory syndrome of a disease, chosen from the group, consisting of autoimmune diseases and immune- related diseases such as asthma, allergy and chronic pulmonary disease; treatment-induced immune-related diseases, such as chemotherapy; infectious diseases, such as viral and bacterial infections; cardiovascular diseases and neurovascular diseases; neuroinflammatory and neurodegenerative diseases; epileptic disorders; affective disorders and psychiatric syndromes; fibrosis; cancer-related disorders; tumour pseudoprogression; cancer and neoplasms; trauma and posttraumatic syndromes; post-organ transplantation syndromes including transplanted organ rejection.
However, the treatment can involve any disease wherein the P2X; receptor activation plays a role and wherein the disease can be treated by a P2X; receptor antagonist.
These diseases are listed in the section ‘diseases involving hyperinflammation’ below.
The hyperinflammatory syndrome preferably includes dyspnoea, in particular, the dyspnoea is associated with a viral infection, bacterial infection, carcinomas, chronic obstructive pulmonary disease (COPD), asthma, allergy, chemotherapy.
The viral infection is particularly caused by a virus, chosen from the group, consisting of Corona, in particular SARS-CoV-2; Influenza; Ebola; Respiratory Syncytial Virus; HIV.
The P2X; receptor antagonist is preferably chosen from the group, consisting of: aminoamide derivatives, in particular lidocaine, bupivacaine, ropivacaine and mepivacaine; antibodies against P2X; receptors, in particular monoclonal antibodies, aminoester derivatives, in particular benzocaine and procaine; adamantane amide derivatives; triazole derivatives; diarylimidazolidine derivatives; pyroglutamic acid amide derivatives; pyrazole acetamide derivatives; dihydrodibenzo [a,g] quinolizinium derivatives; tetrazole derivatives; tyrosine based derivatives; pyrazolodiazepine derivatives; imidazoles derivatives; benzamides derivatives, KN62 analogues and derivatives; adamantane carboxamides; aryl carbohydrazides; cyanoguanidines; aryltetrazoles and aryltriazoles; PPADS tetrasodium salt; brilliant blue G (BBG); oxidised ATP (o- ATP); massadine; stylissadine A and B; P2X; receptor inhibitors C23, C40 and C60; [3H]A-
804598 ([3H]2-cyano-1-[(1S)-1-phenylethyl]-3-quinolin-5-ylguanidine); bicycloheteroaryl compounds. However, additional suitable P2X; receptor antagonists are given in the below section 'P2X; receptor antagonists’. In a very attractive embodiment, the P2X; receptor antagonist comprises lidocaine. Lidocaine has been shown very effective in treatment of hyperinflammation. The lidocaine is preferably administered topically, preferably in the free base form. The administration is preferably in the oral cavity. The lidocaine base is preferably administered in a liquid medium comprising at least
2.5 wiv % of the receptor antagonist, preferably at least 5 w/v%, more preferably at least 10 w/v%. Such a liquid medium can e.g. be ethanol-based.
In another attractive embodiment, the treatment comprises invasive administration of lidocaine in a water soluble salt form, in particular lidocaine-HCI. The lidocaine salt is preferably administered intradermally, subdermally or subcutaneously. The lidocaine salt is preferably administered by continuous intradermal, subdermal or subcutaneous infusion.
The mammalian patient is preferably a human patient, but can be any mammal suffering from a disease mediated by P2X; receptor activation.
The invention will now be further illustrated by way of the following figures and examples. Figure 1 a-f shows six cases with severe COVID-19 treated with subdermal lidocaine. All patients are COVID-19 cases with a positive COVID-19 test. Two patients were treated with mechanical ventilation and extracorporeal membrane oxygenation (ECMO) and 4 patients were treated with mechanical ventilators only. The maximal intravenous lidocaine dose is 0.6 mg/kg/hour and the maximal subdermal lidocaine dose is 1 mg/kg/hour. All patients recovered completely from their illness.
Figure 1a: A 63-year-old male (example 1) with COVID-19-induced ARDS, was admitted to the hospital. The CT scan showed bilateral ground glass opacities. Co-morbidities: COPD, smoking 60 cigarettes per day for more than 40 years. About 40 years before admission the patient suffered from pneumothorax. After admission the clinical condition deteriorated requiring an ICU admission and mechanical ventilation on day 4. On Day 11 continuous intravenous lidocaine of 0.6 mg/kg/hr was initiated but the patient's condition kept worsening with high pulmonary artery pressures and reduced aeration of the lung. On day 19 the continuous intravenous lidocaine of 0.6 mg/kg/hr was changed to continuous subdermal lidocaine of 1 mg/kg/hr. This was followed by improvement of the clinical condition and on day 20 the aeration of the lung was improved but the pulmonary artery pressures remain high. Despite this the P/F ratio was gradually improving and ECMO weaning was done on day 50. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.3 -
0.8%).
Figure 1b: A 68-year-old male with COVID-19-induced ARDS (example 2) admitted to the ICU and required mechanical ventilation. The CT scan showed bilateral ground glass opacities.
Co-morbidity: Asthma. After admission the patient's condition was deteriorating. On Day 5 continuous intravenous lidocaine of 0.6 mg/kg/hr was initiated, but the clinical condition and the P/F ratio kept worsening. On Day 11 the intravenous lidocaine of 0.6 mg/kg/hr was changed to continuous subdermal lidocaine of 1 mg/kg/hr. A few days later this was followed by improvement of the clinical condition and the P/F ratio. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 - 0.6%).
Figure 1c: A 59-year-old male (example 3) with respiratory distress and bilateral ground glass opacities on the CT scan. Co-morbidity: diabetes mellitus and gout. No new ECG changes were observed during treatment with lidocaine. Blood MetHb were within the normal range (0.1 - 0.4%).
Figur1d: A 51-year-old male with fever, dyspnoea and cough due to COVID-19 (example 4). The CT scan showed bilateral ground glass opacities. Co-morbidity: none. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 -
0.3%).
Figure 1e: A 58-year-old male with fever, dyspnoea and cough due to COVID-19 (example 5). The CT scan showed bilateral ground glass opacities. Co-morbidity: Fatty liver. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 - 0.3%).
Figure 1f. A 59-year-old male with fever, dyspnoea and cough due to COVID-19 (example 6). CT scan showed bilateral ground glass opacities. Co-morbidity: Hypertension on medication.
No new ECG changes were observed during treatment with lidocaine. MetHb were within the normal range (0.1 - 0.3%).
Examples 1-6 From April 2020 until end of July 2020, 6 critically ill patients with COVID-19 admitted to the hospital and with intravenous and subdermal continuous infusion of lidocaine. This treatment was initiated on the basis of compassionate use. The concentration of the lidocaine infusion solution is 20 mg/ml (2%). The lidocaine treatment in the first 2 patients was initiated intravenously, the administration route for continuous administration of lidocaine commonly used in daily practice. The dose for intravenous administration is 0.6 mg/kg/hr. Due to the limited efficacy of intravenous lidocaine and based on the hypothesis of selectively targeting the inhibition of the P2X7Rs of the immune cells, the infusion in both patients was converted to subdermal infusion of 1.0 mg/kg/hr after 7 and 6 days, respectively. The other 4 patients were treated with subdermal infusion only. The time course of clinical parameters of these 6 patients is presented in Figures 1a-f.
Patient 1 The first patient, a 63-year-old male (75 kg, 168 cm), developed fever and nausea on March 27, 2020 and three days later he started to cough and became dyspnoeic. After 5 days the PCR COVID-19 test was positive and he was admitted to the hospital with COVID-19-induced ARDS.
Co-morbidities: COPD, smoking 60 cigarettes per day for more than 40 years. About 40 years earlier the patient suffered from pneumothorax. On day 3 the patient deteriorated and was intubated and mechanically ventilated due to poor blood gases. No haemodynamic instability was observed. The CT scan showed bilateral ground glass opacities compatible with ARDS. On day 5 the patient was transferred to the ICU of the University Hospital because of further respiratory deterioration. Prone position mechanical ventilation was initiated due to the progression of the respiratory disease with an extremely low PaO2/FiO2 ratio of 63.3 mm Hg (severe ARDS according to the Berlin definition. The Berlin definition of ARDS includes severe PaO2/FiO:2 ratio £100 mm Hg, moderate PaO:2/FiO2 100 to 200 mm Hg, mild PaO2/FiO2 200 to <300 mm Hg, no ARDS PaO2/FiO2 >300 mm Hg [361]). The initial ventilator settings: APRV, Pnign27 cm HzO, Thign
7.0 s, Pow O cm H20, Tiw 0.32 s. The PaCO: was normal. The echocardiographic estimated pulmonary arterial systolic pressure (PASP) was 80 mm Hg. The Krebs von Lungen 6 (KL-6, a marker for lung fibrosis [362]) plasma level was highly elevated (1299 U/mL; normal value <425 U/mL), CRP was also high (40.4 mg/L; normal value <10 mg/L) and albumin was 2.2 g/dl. The white blood cell count, platelet count and urine production were normal. On day 4 the chest X-ray was not improved. On day 6 the PaO,/FiO; ratio was slightly increased but remained low at 103 mm Hg and the chest X-ray showed progression of the ARDS. ECMO was initiated due to exhausted ventilatory strategy. On day 9 the PaO2/FiO: ratio improved but remained low at around 153 mm Hg but the CRP declined to around 21.8 mg/L. The patient was put on muscle relaxants.
The patient's ARDS status had improved from severe to moderate ARDS. From day 10 until day 30 the ferritin levels were well >1000 ng/ml (>100 ng/dl). From day 11 until day 62 D-Dimer was very high reaching 121.9 nM/L day 14. On day 11 no improvement of the blood gases was observed and it was decided to treat the patient with continuous intravenous lidocaine 0.6 mg/kg/hr. On day 16 CRP showed a progressive decline from 19 (on day 12) to 12.8 (on day 18) and 7.4 (on day 19) but the PaO;/FiO2 ratio remained poor at around 90 mm Hg (severe ARDS according to the Berlin criteria) and the chest X-ray image on day 15, 3 days after the initiation of the intravenous lidocaine infusion, deteriorated dramatically. The lidocaine plasma concentrations were 3.4 ug/ml on day 13 and 5.4 ug/ml on day 14. On day 19 the continuous intravenous lidocaine infusion was replaced by continuous subdermal lidocaine infusion of 1 mg/kg/hr.
Although the PaO./FiO; ratio remained unchanged on day 20 (1 day after the switch to the continuous subdermal lidocaine) the chest X-ray improved clearly. On day 21 the lidocaine plasma concentration was 2.6 g/ml, albumin was 2.5 g/dl. From day 22 on the PaQ./FiOqratio was gradually improving reaching 151 mm Hg on day 34 (moderate ARDS). The KL-6 on day 22 dropped to 458 U/L (this is only slightly above the normal value of <450 U/l). On day 31 The CRP was low at 1 mg/L and the lidocaine plasma concentration was 1.2 g/ml. The muscle relaxants were discontinued. Albumin was 2.3 g/dl. On day 33 the chest X-ray was further improved and the CRP remained low at 5.5 mg/L. The patient was awake and could communicate with the nurses. On day 38 the lidocaine plasma level was 2.3. On Day 43 the PaQ./FiQO; ratio was increased to 214 mm Hg. According to the Berlin definition of ARDS [361], the patient's ARDS status had changed from moderate to mild. Albumin was 2.8 g/dl. On day 50 the patient was weaned from ECMO. On day 51 the patient underwent tracheotomy. Because the clinical condition of the patient was stabilised with a low CRP of 6.3 mg/L on day 55, the continuous subdermal lidocaine was discontinued on day 57. On day 69 he developed pneumothorax requiring pleural drainage. On day 99 he was weaned from the mechanical ventilator and was discharged from the ICU on day 121. The patient received Favipiravir for 14 days. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.3 - 0.8%). The patient was discharged from the hospital on day 187, he went home, he could walk but needed extra oxygen supply of 2L/min. Nine months after admission the patient is doing well and has returned to work.
Patient 2 The second patient is a 68-year-old male with COVID-19-induced ARDS and positive PCR test admitted to the hospital. Co-morbidity: Asthma. The CT scan showed bilateral ground glass opacities. Haemodynamically the patient was stable. On day 2 the respiratory conditions deteriorated, the PaO2/FiO2 ratio is 118 mm Hg (moderate ARDS according to the Berlin ARDS definition [361]). The patient was intubated and required mechanical ventilation. The initial ventilator settings: Pressure control, peak inspiratory pressure 28 cm H2O, PEEP 13 cm H:O, respiratory rate 30/min. CRP was 10.6 mg/L and KL-6 was 486 U/ml. White blood cell count, platelet count and urine production were normal. The ferritin levels remained >1000 ng/ml (100 pg/dl) during the entire ICU stay. Albumin was 2.9 g/dl. The following 3 days the PaO2/FiO: ratio improved to around 150 mm Hg. The PaO:/FiO: ratio dropped from 152 mm Hg on day 5 to 84 mm Hg on Day 6. CRP was increased to 22.9 and the KL-6 was increased to 762 U/ml. The patient was put in prone position and given muscle relaxants. Continuous intravenous lidocaine of 0.6 ml/kg/hr was started. Albumin was 1.8 g/dl. On day 7 the PaO2/FiO: ratio increased to 128 mm Hg, CRP dropped to 10.3 mg/mL and the lidocaine plasma concentration was 2.2 pg/ml. From day 3 until discharge from the ICU D-Dimer values were elevated reaching 75 nM/L on day
14. On day 8, although the PaO2/FiO2 ratio improved from 84 to 125 mm Hg, the mechanical ventilatory strategies were exhausted and the patient was put on ECMO. The KL-6 was increased to 845 U/L and lidocaine plasma level was 2.9 ug/ml. The PaQ./FiO; ratio improved to 238 mm Hg on day 9 but on day 10 a sharp drop of the PaO,/FiO; ratio to 60 mm Hg was observed and CRP was 2.0 mg/ml. The patient's ARDS status had changed from moderate to severe according to the Berlin ARDS criteria (Ranieri et al., Jama 2020, 307 (23), 252 — 2533). Lidocaine treatment was switched from continuous intravenous to continuous subdermal (dosage: 1 mg/kg/hr). On day 14 the lidocaine plasma level was 2.7 pg/ml. KL-6 dropped to 549 U/l. On day 17 the clinical condition of the patient was improving and the PaO2/FiO2 ratio reached 158 mm Hg. The patient was weaned from ECMO. The PaQ,/FiO. ratio improved further reaching 291 mm Hg on day 21 and the patient's ARDS status has changed from moderate to mild ARDS (Ranieri, supra). On Day 22 mechanical ventilation was discontinued and the patient was extubated.
The patient was orientated, no signs of confusion were detected.
Lidocaine treatment was continued until discharge from the ICU on Day 30. The patient received Tocilizumab on day 8 and Favipiravir for 14 days.
No new ECG changes were observed during treatment with lidocaine.
Blood metHb were within the normal range (0.1 - 0.8%). At 3 months after admission the patient is doing well.
Patient 3 The third patient, a 59-year-old male, was admitted to the hospital with respiratory distress and bilateral ground glass opacities on the CT scan.
Co-morbidities: Diabetes mellitus and gout.
The patient required immediate intubation and mechanical ventilation.
The initial ventilator settings: Pressure control, peak inspiratory pressure 30 cm HzO, PEEP 15 cm HO, respiratory rate 25/min.
The PaO2/FiO2 ratio on admission was 160 mm Hg (moderate ARDS according to the Berlin definition [361]), CRP was 39.3 mg/L and KL-6 was 294 U/ml.
White blood cell count was increased 13.10%, platelet count and urine production were normal.
Albumin was 2.1 g/dl.
Haemodynamic parameters were stable.
On the admission day continuous subdermal lidocaine was started at 1 mg/kg/hr.
On day 2 the PaQO,/FiO; ratio improved to 283 mm Hg and the patient's ARDS status had changed from moderate to mild ARDS.
CRP was 41 mg/L, KL-6 was 268 U/I and the lidocaine plasma level was 3.7 ug/ml.
Albumin was 1.7 g/dl.
On day 4 the PaQ2/FiO; ratio was 302 mm Hg, the patient's ARDS status had changed from mild ARDS to no ARDS according to the Berlin ARDS criteria.
On day 5 the PaO:2/FiO2 ratio was improved further to 328 mm Hg and CRP dropped to 16.4 and the patient was extubated.
The patient was orientated, no signs of confusion were detected.
The patient was discharged from the ICU on day 8, CRP was 2.3 mg/ml.
Albumin was 2.5 g/dl.
The patient received Tocilizumab on day 3 and Favipiravir for 15 days.
No new ECG changes were observed during treatment with lidocaine.
Blood MetHb were within the normal range {0.1 - 0.4%). The patient was discharged home on day 20. After 3 months he is doing well.
Patient 4 The fourth patient is a 51-year-old male.
Ten days before admission he developed fever and 2 days before admission dyspnoea and coughing.
On the day of admission, the PCR COVID-19 test was positive.
The CT scan showed bilateral ground glass opacities.
Co-morbidity: none.
The patient was intubated and put on mechanical ventilation on admission.
On day 3 he was transferred to the University Hospital because of deterioration of pulmonary condition.
The initial ventilator settings: Pressure control, peak inspiratory pressure 24 cm HO, PEEP 12 cm H:O, respiratory rate 15/min.
The haemodynamic conditions were stable.
White blood cell count and platelet count were normal.
Albumin was 2.6 g/dl.
Continuous subdermal lidocaine was started immediately.
On day 3 the PaO2/FiO: ratio was 214 (moderate ARDS according to the Berlin definition [361]). KL-6 was 177 U/L and CRP was 17.4 mg/L. On day 5 the PaO:/FiO2 ratio was increased to 382 (the patient's ARDS status had changed from mild ARDS to no ARDS) and lidocaine plasma concentration was 5.2 ug/ml. CRP was 27.3mg/L. Lidocaine plasma levels on day 3 and 4 were 3.4 and 4.2 pg/ml, respectively. KL-6 was 163 U/L. The patient was extubated. The patient was orientated, no signs of confusion were detected. The patient was discharged from the ICU on day 8, the CRP was 9.3 mg/L. The patient received Favipiravir for 14 days. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 - 0.3%). He was discharged home on day 28. At 3 months he is doing well and has returned to work.
Patient 5 The fifth patient is a 58-year-old male. Nine days before admission he developed a sore throat. A day later he developed fever. Two days before admission he started coughing and was dyspnoeic. On the day of admission the PCR COVID-19 test was positive. The CT scan showed bilateral ground glass opacities. Co-morbidity: Fatty liver. The patient was initially admitted to the hospital ward. On day 3 the patient deteriorated and had to be intubated and put on mechanical ventilation. On day 4 the patient was transferred to the University Hospital due to deterioration of the pulmonary condition. The initial ventilator settings: Pressure control, peak inspiratory pressure 27 cm H20, PEEP 12 cm H20, respiratory rate 25/min. PaO2/FiO: ratio was 188 (moderate ARDS according to the Berlin definition). Haemodynamic parameters were stable and CRP was 12.9 mg/ml. White blood cell count was increased (14.4.10%L) but platelet count was normal. KL-6 was 330 U/L. Continuous subdermal lidocaine was started at 1 mg/kg/hr. Albumin was 2.8 g/dl. On day 5 the PaO./FiO; ratio was unchanged, CRP was 10.4 mg/L and the lidocaine plasma level was 4 ug/ml. On day 6 the lidocaine plasma level was 3.2 pg/ml. KL-6 remained stable at 400 U/L. Albumin was 2.3 g/dl. On day 10 the respiratory insufficiency had cleared, although the PaO2/FiO2 ratio remained 184 the CRP dropped to 2.4 mg/L and KL-6 was 322 U/L. The patient was extubated and he was orientated, no signs of confusion were detected. On day 14 the patient was discharged from the ICU. The patient received Tocilizumab on day 7 and Favipiravir for 10 days. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 - 0.3%). On day 20 the patient was discharged home and is doing well at 3 months after admission. Patient 6 The sixth patient is a 59-year-old male with fever, dyspnoea and cough due to COVID-19. CT scan showed bilateral ground glass opacities. Co-morbidity: Hypertension on medication. The patient was admitted to the general ward. KL-6 233 U/L, white blood cell count and platelet count were normal. Albumin was 3.6 g/dl. On day 3 there is a deterioration of the respiratory function necessitating a transfer to the ICU and mechanical ventilation. The initial ventilator settings:
Pressure control, peak inspiratory pressure 22 cm HO, PEEP 10 cm H:O, respiratory rate 20/min. Continuous subdermal lidocaine of 1 mg/kg/hr was initiated after admission to the ICU. Haemodynamic parameters were stable. CRP was 6.3 mg/L, KL-6 was 263 U/L. On day 4 a progressive respiratory failure occurred requiring intubation and mechanical ventilation. Pa0/FiO; ratio was 218 mm Hg, the haemodynamic parameters remained stable. CRP was 6.3 mg/L, the white blood count and platelet count were normal. Lidocaine plasma level was 4.6 pg/ml. On day 5 the PaO:2/FiO: ratio dropped further to 164 mm Hg. Lidocaine plasma level was
3.4 pg/ml. Albumin was 3.2 g/dl. On day 9 the clinical condition of the patient improved. The ventilator settings could be decreased, the PaO2/FiO2 ratio remained 207 mm Hg during the weaning period, CRP was 0.7 mg/L. On day 10 the patient was extubated, he was orientated, no signs of confusion were detected. On day 13 the patient was discharged from the ICU. Tocilizumab was given on day 4. The patient received Favipiravir for 11 days. No new ECG changes were observed during treatment with lidocaine. Blood metHb were within the normal range (0.1 - 0.3%). He was discharged from the hospital on day 20 and at 3 months after admission he is doing well, played golf and has returned to work. Example 7 — COVID-19 On March 24, 2020, a 49-year-old female developed cough, dyspnoea, myalgia, pain in hip and groin, shivers but no fever. A COVID-19 test was not performed. This occurred one week after closely working together with a colleague who developed proven severe COVID-19 requiring hospital admission. Treatment with lidocaine was started 4 hours after the symptoms developed. After the initiation of continuous subdermal (superficial subcutaneous) infusion of lidocaine of 0.5 mg/kg/hr the symptoms subsided gradually and after a few hours she was practically free of symptoms. After 12 hours the lidocaine was discontinued. After a few hours the symptoms relapsed and subsided again after reinitiating the continuous subcutaneous lidocaine infusion. The lidocaine treatment lasted for 3 days. On April 1, 2020 her symptoms returned and lidocaine subdermal infusion was reinitiated. After a few days the clinical condition improved and lidocaine treatment was discontinued. The patient could work from home but experienced fatigue. After 4 weeks she recovered completely from all symptoms. On July 12, 2020, (day 1) the patient developed new symptoms comprising shivers, joint pain (knees, hips, shoulders) and conjunctivitis of the left eye. She reported sick at work. The infection probably occurred on July 1, 2020. On that day she met many people during her work at the office and in the evening she went for a drink in a bar with a few colleagues. The infection occurred despite social distancing and hand sanitising. At that time no one wore a face mask. She was treated with the subdermal infusion of lidocaine (0.63 mg/kg/hr). Her symptoms improved within a few hours. On July 15 (day 4), the lidocaine infusion was stopped for several hours, she went to the GP for a COVID-19 test. On July 21 (day 10), 2020, the IgA antibody test for COVID-19 result was positive. After a week the lidocaine infusion was discontinued during daytime due to the infusion system and syringe pump being a serious obstacle for daily activity.
On day 11 the symptoms increased: Prickling sensation in the lungs, chest pain, shivers, dyspnoea, frequent yawning and dizziness. The lidocaine infusion was given 24 hours per day.
The symptoms improved gradually.
On day 12 she developed increasing chest pain, shivers and pain in the proximal muscles and joints. Temperature 35.9 centigrade, SpO: (oxygen saturation) 86%, HR (heart rate) 70/min, BP (blood pressure) 110/70 mm Hg. Bronchial breath sounds were heard over the left inferior lobe of the lung and to a much lesser extent over the right inferior lobe. Subdermal lidocaine infusion (0.63 mg/kg/hr) still running and in addition she was treated with amoxicilline3 x 500 mg/day under suspicion of secondary bacterial pneumonia. She fell asleep and the following day all symptoms were improved significantly and the infusion was continued uninterruptedly.
On day 18 the subdermal lidocaine infusion was discontinued because the patient's daily activities were seriously hampered by the infusion system and syringe pump. A few hours later a clinical deterioration occurred with extreme fatigue, chest pain and pain in the right upper arm. The treatment was converted to transdermal lidocaine cream. Formulation: 2,5% alpha-terpineol, 20% lidocaine, 10% castor oil , 1% Polysorbate 20; 0,5% Carbopol and 56% water (lidocaine cream 200 mg/ml). As much as 400 mg cream is applied over the skin and covered by Tegaderm transparent wound dressing. But after 1 hour the symptoms did not improve and subdermal lidocaine infusion of 40 mg/hour was initiated. After 1 hour the clinical improvement was noticeable. But after 6 hours the infusion rate was increased to 63 mg/hour (1 mg/kg/hr) due to persistent symptoms. Thereafter the symptoms disappeared with the exception of the pain in the upper arm.
Day 19, the pain in the right arm remained for several weeks. The pain is mostly felt in the upper arm muscles and some times in the shoulder, elbow and lower arm. This lead to a serious frozen shoulder syndrome.
Day 22, a period with excessive yawning and body temperature (orally measured) of 34°C.
On day 23 the patient developed undulating shivers with coughing, dyspnoea, prickling sensations of the lungs, chest pain, sore throat, headaches, yawning, conjunctivitis of the left eye and joint pain. Lidocaine infusion reduced the symptoms to tolerable levels.
On day 24 the patient has had enough of the infusion and the lidocaine infusion was converted to transdermal lidocaine cream 20% of 400 mg applied on a surface of 2 x 50 cm? and covered with Tegaderm. The cream was almost completely absorbed after 6 hours and each time was replaced with a new dose.
Day 25, the Tegaderm wound dressing was damaged during the night and part of the dose of the cream was lost. She woke up with sternal pain, pain in the area of the right dorsal Latissimus muscle with a feeling of malaise. SpO:2 98%, HR 74/min, BP 114/71, temperature 35.3 degrees centigrade. A new cream dose was applied and an extra 100 mg cream was rubbed into the skin as a bolus dose. After 1 hour the clinical condition improved. The lidocaine 400 mg cream was set to be administered every 6 hours.
Day 26, it appeared that the cream, applied on the abdomen wall, was not absorbed well by the skin during the night. Her symptoms returned and a new dose of lidocaine cream, applied on the upper leg, reduced the symptoms within 1 hour. At 16:00 the cream was completely absorbed and the symptoms returned again. New lidocaine cream dose is effective once more.
Day 27, the resorption of the lidocaine cream applied on the arms and abdominal wall is much less than on the inner upper legs.
On day 32, 07:30, the absorption of the lidocaine cream was less than 80%. Her clinical condition was worse, the SpO; dipped regularly to 90%. Beclomethasone nose spray 2 x 200 ug was given. She needed regular sigh and coughing to keep the SpO: above 92%. An extra 100 mg cream was rubbed into the skin as a bolus dose resulting in an improvement of the symptoms within 30 minutes. At 12:00 left knee became extremely painful and was treated again with a new cream dose and an extra 100 mg cream was rubbed into the skin as a bolus dose again resulting in the improvement of the symptoms. The total daily dose of lidocaine cream was 2000 mg.
The night of day 32 to 33 was, apart from the painful upper right arm, without any symptoms. The first night without symptoms since day 1 (July 12, 2020).
On day 33 at 11:10 the patient experienced a sudden and extreme pain between her scapulae. She was dyspnoeic, she hyperventilated and her face went pale. SpO. 97%, HR 66/min, BP 112/60 mm Hg, temperature 35.3 degrees centigrade. Analysis at the hospital revealed a normal lung X-ray, normal metHb and normal d-dimer. Further routine laboratory results were also normal.
On day 35, apart from fatigue and pain in the right upper arm the patient had no other symptoms. The lidocaine cream was tapered down to 4 x 100 mg/d without Tegaderm covering.
Day 46, 03:30, SpO: alarm at 86%, HR 55/min, BP 115/70 mm Hg, prickling sensations of the lung, left conjunctivitis. The lidocaine cream was removed and continuous subdermal infusion of 0.63 mg/kg/hr lidocaine was initiated. The symptoms improved within 30 minutes. The painful right upper arm remained unchanged.
After 1 week the lidocaine infusion was discontinued.
On day 61 at 04:20 a new episode with prickling sensations of the lung, left conjunctivitis and increased painful upper right arm developed. Continuous subdermal infusion of 0.63 mg/kg/hr lidocaine was initiated and the symptoms subsided. Lidocaine infusion was discontinued after 8 hours. At 19:40 after dinner, the patient developed shivers, was feeling unwell and had to go to bed. Temperature 34.1, BP 96/60 mm Hg, SpO: 100%, HR 86/min. Continuous subdermal infusion of 0.63 mg/kg/hr lidocaine was re-initiated with 400 ug of beclomethasone nose spray and oral hydrocortisone. The symptoms quickly subsided.
On day 66 she felt good apart from the painful right upper arm. The pain in the upper arm is fluctuating in intensity and sometimes migrates to the shoulder or elbow. The general practitioner was not convinced that this had anything to do with COVID-19. The lidocaine infusion was discontinued.
On day 67, she made a trip by car, as a passenger, to visit friends in a place 200 km from home.
Day 85, after almost 3 weeks with no symptoms other than a fluctuating pain in the upper arm, the prickling sensations of the lung returned. SpO2 96%, HR 90/min, BP 95/68 mm Hg, temperature 33.5 degrees centigrade. Sublingual lidocaine 3 x 100 mg/d was initiated. The lidocaine was kept in the mouth for 15 minutes and then swallowed. Inhalation of the lidocaine solution was avoided. Formulation of the lidocaine: Xylocaine 5 g in 50 ml (10% solution, 100 mg/ml}, ethanol 96%, polyethylene glycol 400, banana essence, and purified water.
From day 85 on, after the initiation of the sublingual lidocaine treatment, the patient experienced prickling sensations and oppressive feeling on her chest in the morning before she took the medication and at the end of the day before taking the sublingual lidocaine. These symptoms disappeared 20 minutes after the administration of sublingual lidocaine. This occurred repeatedly almost every day.
Example 8 - COVID-19 On December 20, 2020, a 46-year-old male developed a runny nose, mild headache, stiffness and severe pain of the neck and right shoulder, loss of smell and strongly diminished taste. He felt washed-out and lethargic. The patient had had contact with a COVID-19 patient 5 days earlier. On day 2 after the initiation of the symptoms the PCR swab test for COVID-19 was positive. The following days symptoms were progressive.
On day 5 the patient received 5 x 60 mg lidocaine using a metered dose. The drug was administered sublingually and kept in the mouth for 15 minutes. Thereafter, the lidocaine solution was swallowed. Inhalation of the lidocaine was avoided. Formulation of the lidocaine: Xylocaine 5 gin 50 ml (10% solution, 100 mg/ml), ethanol 96%, polyethylene glycol 400, banana essence, and purified water. On day 6 (24 hours after the initiation of the treatment) the runny nose, headache, neck pain and neck stiffness disappeared. On day 6 the patient received lidocaine 4 x 60 mg/d and from day 7 on the patient was treated with lidocaine 3 x 60 mg/d. On day 9 the patient is much less lethargic, felt to have more energy and started cleaning his house thoroughly. He reported dry mouth feelings for the first time. On day 8 all symptoms, with the exception of dry mouth feelings and mild fatigue, disappeared completely. On day 11 the patient took a 1 hour walk and felt good.
Example 9 — ARDS by Staphylococcus sepsis An example in off-label use of lidocaine as the ultimate drug {ultimum remedium) to treat a patient suffering from severe ARDS. A 43-year-old female was admitted to an ICU in the Hague region at the end of 2019. She developed severe ARDS from staphylococcus sepsis. This sepsis developed after intravenous administration of contrast fluid for an MRI image. The patient was put on mechanical ventilation. Despite adequate antibiotic treatment, the ARDS and sepsis deteriorated further leading to insufficient oxygenation under mechanical ventilation with haemodynamic instability requiring very high doses of nor-adrenaline and vasopressin. She was connected to ECMO (extracorporeal membrane oxygenation) and transferred to the University Hospital Rotterdam. It was planned to keep the patient on the ECMO, operatively remove the lungs and to treat the patient with antibiotics for 2 months to clear the thorax cavities from microbes. This procedure would then be followed by a lung transplantation. The haemodynamic instability and the poor oxygenation, even with ECMO therapy, motivated the intensivist in charge to treat the patient with continuous low dose lidocaine aiming at the inhibition of the P2X7R. Within
1.5 hours after the initiation of the continuous lidocaine infusion of 1 mg/kg/hour, the patient's condition stabilised. Over the following days the noradrenalin and vasopressin medication could be tapered down and after several days the ECMO was disconnected because oxygenation with regular ventilation was restored. Needless to say that the planned lung transplantation was cancelled. After 1.5 months, the patient was weaned from the ventilator and was transferred to the ward. The patient received lidocaine for a period of 2 weeks.
Example 10 — Polymyalgia rheumatica In January 2012 a 59-year-old man suffered from progressive muscle pains, initially referred to as statin related. Could hardly turn over in bed. The patient experienced varying loss of strength, weight remained stable 54 kg. No fever. In addition, the patient reported fatigue and general malaise. No familial skin or muscle disorders, but asthma and CVD. Eighteen months previously, the patient had a tick bite and developed erythema with erythema chronicum migrans (ECM). On the suspicion of an infection with Borrelia Burgdorferi the patient was treated with amoxicillin for 4 weeks. Laboratory results revealed an increasing erythrocyte sedimentation rate of 47 mm/hr (November 1, 2012) and 85 mm/hr (February 2013). Chest X-ray and CT scan of the chest and abdomen were unremarkable. The diagnosis was polymyalgia rheumatica. The patient was initially treated with high dose corticosteroids. This medication was tapered down and discontinued after 6 months.
In September 2019 the symptoms recurred. The patient was initially treated with nocturnal continuous subdermal lidocaine infusion of 0.5 mg/kg/hr for 8 hours with a frequency of 2 x/week. The symptoms disappeared after the first treatment. After a few months the lidocaine subdermal infusion was replaced with transdermal 5% lidocaine ointment 300 mg twice a day. This therapy was ineffective. On September 10, 2020 the patient started to use sublingual lidocaine 2 x 60 mg/d. The lidocaine was kept in the mouth for 15 minutes and then swallowed. Inhalation of the lidocaine solution was avoided. Formulation of the lidocaine: Xylocaine 5 g in 50 ml (10% solution, 100 mg/ml), ethanol 96%, polyethylene glycol 400, banana essence, and purified water. The symptoms disappeared within 1 hour and (during treatment) did not return until the end of the follow-up on January 2, 2021. Example 11 — Psoriatic arthritis A 60-year-old female was presented with cutaneous herpes zoster infection of her back extending to the abdomen wall.
Four years earlier she developed psoriasis.
In the previous year she developed progressive rheumatoid arthritis and lichen planus.
The symptoms were progressive and since a few weeks she was not able to cook her meals.
She could barely get dressed and get undressed.
She had been treated with corticosteroids and methotrexate.
In the beginning of July 2020 a very painful herpes zoster infection of the trunk emerged and the patient was treated with morphine.
Morphine did not subside the pain but she developed obstipation.
This is the situation when she was presented to us.
On August 8, 2020 she started with our treatment consisting of lidocaine cream 10%. Formulation: 2,5% alpha-terpineol, 10% lidocaine, 10% castor oil , 1% Polysorbate 20; 0,5% Carbopol and 66% water (lidocaine cream 100 mg/ml). Lidocaine cream dosage was 2 x 200 mg/day applied over the skin of her forearm and covered by Tegaderm transparent wound dressing.
Every day, the location of the applied cream was alternately on the left and right forearm.
The morphine treatment was tapered down.
Because the Tegaderm covering damages the skin she changed to cling film covering wrapped with elastic bandage.
The obstipation disappeared within 2 days, the herpes zoster infection was rapidly declining and after 14 days her sense of well-being increased from 3 to 7.5 on a scale of 10. She could perform her daily activities at home normally.
After 4 weeks she could easily do some gardening and her capacity to perform her daily activities returned to normal.
Example 12 — Sondylattosis A 62-year-old patient from Italy was presented with an intractable progressive and debilitating degenerative disease of the vertebrae (L2-L3, L3-L4 and L4-L5). He suffered severe pain, could hardly walk, could no longer do his work as a crane operator and was no longer able to practice his hobby (repairing racing motorcycles). The symptoms had been progressive for 20 years.
Pain killers had no effect.
December 2019 he received 2 x 1400 mg/d lidocaine patches.
To prevent skin irritations, the patches were administered on alternate locations on the skin.
According to the patient, 70% and 90% of the symptoms have disappeared after 4 days and 2 weeks, respectively.
He has little pain, went back to work and was able to pick up his hobby again.
Example 13 - Chronic interstitial cystitis (chronic inflammatory bladder condition) An 88-year female, co-morbidity hypertension, glaucoma, diabetes type II with renal dysfunction, chronic inflammation of the uterus and the bladder.
The past 2 years she complained about intractable progressive extreme pain of the bladder especially during micturition.
Oxycodone (morphine analogue), paracetamol and antibiotic treatment had no effect on her symptoms. Treatment with continuous subcutaneous lidocaine of 1 mg/kg/hr reduced her symptoms gradually and after 2 days she was practically free of symptoms. After 1 week the treatment was discontinued due to problems with the supply of the drug and her symptoms reoccurred. After restarting the lidocaine infusion her symptoms disappeared once again.
Example 14 - Knee arthrosis A 43-year old female suffered from gonarthrosis (knee osteoarthritis) of the right knee. There is hydrops of the right knee and is extremely painful. Her maximal walking range is 100 m before the pain forced her to stop walking and to sit down. She could barely climb the stairs at home and she was not able to ride her bicycle. Because there is a defect in the ventral part of the femorotibial cartilage tissue she was rejected for a knee extraction therapy. On January 25, 2020, she received treatment with 2 x 1400 mg/d lidocaine patches. Within 6 weeks the hydrops of the right knee and the pain improved, her walking range was extended to >500 m, she could climb the stairs and after 8 weeks she could ride her bicycle.
Example 15 - Multiple sclerosis A 46-year-old patient was diagnosed with relapsing-remitting multiple sclerosis (RRMS) in
2005. In 2005-2006 she was treated with Avonex. In January 2020 she was presented with fatigue. She had to take a rest between 10: AM and 14:00 because of fatigue. Her neurological symptoms consist of paraesthesia of the right arm. We treated her with 1 x 700 mg/48 hours lidocaine patches. After 4 weeks her fatigue improved, she could skip resting during the days and she felt to have more energy for her daily activities.
Example 16 - Advanced cervix carcinoma and renal dysfunction.
A 65-year-old female was diagnosed with advanced cervix carcinoma in July 2018. The tumour mass blocked both ureters and caused bilateral hydronephrosis. The hydronephrosis was drained successfully and the patient was treated with paclitaxel and bevacizumab. In October 2018 renal function impairment developed. In August 2019 the CT scan of the chest and abdomen revealed a large tumour mass in the pelvis cavity, multiple para-iliac lymph node metastasis and a mass surrounding the pancreas. She developed cancer-related ascites. The conclusion was that the tumour growth was progressive. But because of the progressive impairment of the renal function, the intended therapy with carboplatin and gemcitabine was postponed.
On September 15, 2019 the patient started to receive continuous subdermal lidocaine infusion of 1 mg/kg/hr. The renal function improved. After 6 weeks the renal function improvement was such that the patient could be treated with carboplatin and gemcitabine. Five months later, in February 2020, the tumour growth was stabilised and the chemotherapy treatment could be discontinued. At that time she was still on lidocaine infusion.
Example 17 — diverse cancer 10 patients: Prostate cancer (2 patients), exocrine pancreatic cancer (4 patients), colon cancer (2 patients), cervical carcinoma (1 patient) and breast cancer (1 patient). All patients had no more options for further cancer treatment left and all patients were treated with palliative opiates. We prescribed the patients continuous subcutaneous lidocaine 1 mg/kg/hr instead of morphine.
In 7 patients the relieve of pain, nausea, and/or extreme fatigue was achieved within 2 hours and in the remaining patients within 48 hours. This improved further in the following week and stabilised for weeks.
For example, an 81-year old patient with a terminal metastatic colon cancer with much pain, discomfort and malignant ascites, improved so much that after 3 weeks he could perform with his rock band in a wheel chair Sickness impact profile for disability (SIP68) was applied for inventory data. Before treatment: SIP68: 48 (range 18 - 68). After 3 days: SIP68: 33 (range 12-58), all patients were improved. After 7 days: SIP68: 28 (range 12 - 56), all patients were improved. Section P2X7 receptor antagonists Monoclonal antibodies specific against P2X7R Chemical compounds Amides derivatives
1. Oxo-proline amide derivatives as described in KR101398264B1
2. Amino amide group: Lidocaine, Articaine, Bupivacaine, Cinchocaine (Dibucaine), Etidocaine, Levobupivacaine, Lidocaine (Lignocaine), Mepivacaine, Prilocaine, Ropivacaine, Trimecaine Amino ester derivatives Procaine, Benzocaine, Chloroprocaine, Cocaine, Cyclomethycaine, Dimethocaine (Larocaine), Piperocaine, Propoxycaine, Proparacaine, Tetracaine (Amethocaine) Phenyl-Substituted 5,6-Dihydro-[1,2,4]triazolo[4,3-a]pyrazine P2X7 Antagonists Bicycloheteroaryl compounds as p2x7 modulators and uses thereof (WO2007/109192) P2X7R inhibitors used in clinical trials Three P2X7R antagonists have been tested in human: CE-224,535 500 (Pfizer), AZD9056 (Astra-Zeneca) and JNJ-54175446 (Johnson and Johnson).
P2X7R inhibitors published by Mehta N, et al. Bioorg Med Chem, 201422 (1) 54 — 88
1. Existing P2XR inhibitors not selective for P2X7R only: PPADS tetrasodium salt, brilliant blue G (BBG), oxidised ATP (0-ATP)
2. Existing specific 2X7R inhibitors: KN-62, AZ9056, A-740003, A-438079, GSK314181A, A- 804598, A-839977 and AZ-116453743.
3. Parent scaffold based classification, 181 compounds: Adamantane amide derivatives, Triazole derivatives, Diarylimidazolidine derivatives, Pyroglutamic acid amide derivatives, Pyrazole acetamide derivatives, Dihydrodibenzo [a,g] quinolizinium derivatives, Tetrazole derivatives, Tyrosine based derivatives, Pyrazolodiazepine derivatives, Imidazoles derivatives, Benzamides derivatives, KN62 analogs derivatives, Natural antagonists of P2X7
4. Three P2X7R inhibitors from natural product extracts: Massadine, Stylissadine A and Stylissadine B. P2X7R inhibitors published by Caseley EA, et al. Biochem Pharmacol 2016 (116) 130 — 139
1. 73 top-ranked compounds after virtual screening of approximately100,000 structurally diverse compounds against the ATP-binding pocket in the hP2X7R: C1 until C73.
2. Three of these compounds appeared to effectively inhibit transmembrane currents due to P2X7R activation and macropore forming (YO-PRO-1 uptake): C23, C40 and C60.
P2X7R inhibitors published by Bin Dayel A, et al. Mol Pharmacol 2019, 96 (3) 355 — 363 AZ11645373, brilliant blue G, KN-62, calmidazolium and ZINC58368839. P2X7R inhibitors published by North RA, et al. Physiol Rev 2002, 82, (4), 1013-67.
1. Existing P2XR inhibitors not selective for P2X7R only: BBG, PPADS, suramin, o-ATP.
2. Existing specific 2X7R inhibitors: A-438079, A-804598, A-740003.0, KN-62, AZ10606120, AZ11645373, GW791343 and JNJ47965567 P2X7R inhibitors published by Sluyter, Adv Exp Med Biol -Prot Rev 2017, (19) — 17 - 53
1. Existing P2XR inhibitors not selective for P2X7R only: BBG, o-ATP, PPNDS, PPADS, MRS2159, NF279, NF449.
2. Existing specific 2X7R inhibitors: AACBA, AstraZeneca, A-438079, A-804598, A-
740003.0 and KN-62. P2X7R inhibitors published by Carroll, et al. Purinergic signal 2009, 5, (1) 63 — 73 57 Compounds: Adamantane carboxamides, Aryl carbohydrazides, Cyanoguanidines, Aryltetrazoles/aryltriazoles
P2X7R inhibitor published by Donnelly-Roberts DL, et al., Neuropharmacology 2009, 56, (1), 223-9. [3H]A-804598 ([3H]2-cyano-1-[(1S)-1-phenylethyl]-3-quinolin-5-ylguanidine) P2X7R inhibitor published by Ruiz-Ruiz q, et al., Front Mol Neurosci 2020, 13, 93. AZ11645373, AZD-9056, A-438079, A740003, CE-224,535, GSK-1482160, JNJ-47965567, A-804598, 2, JNJ-54175448, JNJ-55308942 Section Diseases involving hyperinflammation — involving the activation of p2X; receptor of the immune system
1. Autoimmune diseases and immune-related diseases
2. Treatment-induced immune-related diseases
3. Infectious diseases
4. Cardiovascular diseases and neurovascular diseases
5. Neuroinflammatory and neurodegenerative diseases
6. Epileptic disorders
7. Affective disorders and psychiatric syndromes
8. Fibrosis
9. Cancer-related disorders
10. Cancer and neoplasms
11. Trauma and posttraumatic syndromes
12. Post-organ transplantation syndromes including transplanted organ rejection Autoimmune diseases and immune-related diseases Primary immunodeficiency Systemic inflammatory diseases
1. Systemic syndrome in advance cancers (SIRS).
2. Sepsis a. Bacterial sepsis: Pseudomonas spp., Staphylococcus spp., Streptococcus spp. b. Viral sepsis and ARDS: Influenza A virus, SARS, MERS, COVID-18, etc. c. Overwhelming post-splenectomy sepsis: Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis d. Babesiosis: Babesia microti (U.S.), B. divergens (Europe) e. Meningococcemia with sepsis and meningitis: N. meningitides f. Rocky Mountain spotted fever (RMSF): Rickettsia rickettsii g. Purpura fulminans: S. pneumoniae, H. influenzae, N. meningitidis h. Erythroderma, toxic shock syndrome: Group A Streptococcus, Staphylococcus aureus i. Necrotizing fasciitis: Group A Streptococcus, mixed aerobic/anaerobic flora, CA- MRSA® j. Clostridial myonecrosis: Clostridium perfringens k. Gas gangrene
3. Hyperinflammation and cytokine storm
4. Anaphylactic reaction including shock
5. Systemic allergic reactions
6. Systemic reactions (SIRS) following trauma
7. Acute post-operative (post-transplantation) inflammation and SIRS Endocrine diseases
1. Type | and type II diabetes
2. Addison’s disease
3. Autoimmune polyendocrine syndrome (APS) type 1, 2 and 3
4. Autoimmune pancreatitis (AIP)
5. Autoimmune thyroiditis
6. Ord's thyroiditis
7. Grave's disease
8. Hashimoto's disease
9. Autoimmune oophoritis
10. Endometriosis
11. Autoimmune orchitis
12. Sjögren's syndrome
13. Osteoporosis
14. Paget's disease Connective tissue diseases
1. Mixed connective tissue disease
2. Undifferentiated connective tissue disease
3. Adiposis dolorosa
4. Systemic lupus erythematosus (SLE)
5. Drug-induced lupus
8. Adult-onset Still's disease
7. CREST syndrome
8. Enteritis-related arthritis
9. Eosinophilic fasciitis
10. Felty syndrome
11. IgG4-related disease
12. Parry-Romberg syndrome
13. Parsonage-Turner syndrome
14. Sarcoidosis
15. Schnitzler syndrome
16. Undifferentiated connective tissue disease (UCTD) Eye diseases
1. Diabetic retinopathy
2. Autoimmune retinopathy
3. Autoimmune uveitis
4. Intermediate uveitis
5. Dry and wet Age-related Macular Degeneration (AMD)
6. Retinitis Pigmentosa (RP)
7. Ligneous conjunctivitis
8. Mooren's ulcer
9. Scleritis
10. Sympathetic ophthalmia Ear diseases Autoimmune inner ear disease (AIED) Pulmonary diseases
1. Asthma
2. Allergic rhinitis
3. Chronic obstructive pulmonary disease (COPD)
4. Autoimmune inner ear disease (AIED) Gastrointestinal diseases
5. Drug-induced liver diseases
6. Autoimmune hepatitis
7. Inflammatory bowel syndrome
8. Crohn's disease
9. Ulcerative colitis
10. Irritable bowel syndrome
11. Microscopic colitis
12. Autoimmune enteropathy
13. Coeliac disease
14. Gluten intolerance
15. Lactose intolerance
16. Plummer-Vinson syndrome
17. Achalasia
18. Idiopathic peritonitis Diseases of muscles, bone and skin
1. Skin immune response following insect bites and stings
2. Contact dermatitis
3. Polymyositis
4. Myositis
5. Dermatomyositis
6. Dermatitis of different causes
7. Inclusion body myositis
8. Fibromyalgia
9. Systemic scleroderma
10. Psoriasis
11. Alopecia areata
12. Autoimmune angioedema
13. Autoimmune progesterone dermatitis
14. Autoimmune urticarial
15. Bullous pemphigoid
16. Cicatricial pemphigoid
17. Gestational pemphigoid
18. Dermatitis herpetiformis
19. Discoid lupus erythematosus
20. Epidermolysis bullosa acquisita
21. Erythema nodosum
22. Hidradenitis suppurativa
23. Lichen planus
24. Lichen sclerosus
25. Linear IgA disease (LAD)
26. Morphea
27. Pemphigus vulgaris
28. Pityriasis a. Pityriasis alba b. Pityriasis lichenoides chronica c. Pityriasis rosea d. Pityriasis circinata e. Pityriasis rubra pilaris f. Pityriasis versicolor g. Dandruff, historically called Pityriasis capitis h. Pityriasis amiantacea i. Pityriasis lichenoides et varioliformis acuta (PLEVA) j. Mucha-Habermann disease
29. Vitiligo
30. Angioedema a. Acquired angioedema b. Hereditary angioedema c. Antineurotic oedema (Quincke’s oedema)
31. Eczema
32. Rheumatoid arthritis
33. Chronic inflammation of the knee joints, hip joints, spine joints, etc.
34. Chronic spondylarthrosis
35. Chronic osteochondritis and osteoarthritis
36. Juvenile arthritis
37. Ankylosing spondylitis
38. Psoriatic arthritis
39. Palindromic rheumatism
40. Relapsing polychondritis
41. Polymyalgia rheumatica
42. Antisynthetase syndrome Urogenital diseases
1. Chronic interstitial cystitis
2. Recurrent cystitis
3. Drug-induced nephropathies
4. Diabetic nephropathy
5. Nephrotic syndrome
6. The nephritic syndrome
7. Rapidly progressive glomerulonephritis
8. Acute renal failure
9. Secondary renal dysfunction Treatment-induced immune-related diseases
1. Radiation-induced encephalopathy
2. Chemotherapy-related kidney injury a. Complex renal cysts b. Interstitial nephritis c. Renal papillary necrosis d. Renal infarction e. Acute tubular necrosis
3. Chemotherapy-related bladder injury a. Chemotherapy-induced cystitis b. Haemorrhagic cystitis
4. Chemotherapy-related gastrointestinal injury a. Stomatitis b. Pharyngitis c. Oesophagopharyngitis d. Mucositis e. Oral and anal inflammation or ulceration f. Bowel necrosis g. Gastrointestinal ulceration h. Enteritis i. Pancreatitis j. Acute hepatitis Infectious diseases
1. Viral disease a. Hepatitis A, B, C and D (HAV, HBV, HCV and HDV) b. Human retroviruses c. Human immunodeficiency virus (HIV) d. HTLV-1 e. Abelson murine leukaemia virus f. Rous sarcoma virus g. Zika virus (ZiKV) h. Influenza A and B virus (IAV and IBV) i. Coronaviruses e MERS e SARS e SARS-CoV-2 j. Rhinoviruses k. Human respiratory syncytial virus I. Adenoviruses m. Enteroviruses n. Human metapneumoviruses
0. Herpes simplex or zoster encephalitis p. Herpes simplex or zoster dermatitis gq. Herpesvirus type 6, 7 and 8 r. Dengue virus s. West Nile virus t. Ebolavirus and Marburgvirus Infections u. Hendra virus v. Nipa virus w. Human papilloma virus (HPV) X. Epstein-Barr virus (EBV), Infectious mononucleosis y. Rubeola: Measles virus z. Rubella aa. Mumps bb. Smallpox cc. Chickenpox dd. Yellow fever ee. Viral myocarditis ff. Viral haemorrhagic fever gg. Rabies hh. Hand-foot-and-mouth disease ii. Orf Parapoxvirus jl. Molluscum contagiosum Scrub typhus kk. Norovirus Il. Cytomegalovirus (CMV) mm. Condyloma acuminatum nn. Parvovirus
00. Arthropod-borne and rodent-borne virus infections
2. Bacterial infections a. Cat-scratch disease b. Tularemia c. Donovanosis d. Nocardiosis e. Actinomycosis and Whipple's Disease f. Typhoid g. Leptospirosis h. Q fever i. Brucellosis j. Melioidosis k. Echinococcal (hydatid) disease I. Chronic osteomyelitis m. Lyme disease (Borrelia burgdorferi) n. Tick-borne spotted fevers o. Tuberculosis p. Buruli ulcer: Mycobacterium ulcerans g. Cutaneous tuberculosis: M. tuberculosis r. Leprosy: M. leprae s. Helicobacter pylori t. Schistosomiasis u. Histoplasmosis v. Entamoeba histolytica w. Giardiasis x. Filariasis y. Visceral larva migrans z. Anthrax: Bacillus anthracis aa. Ulceroglandular tularemia” Francisella tularensis bb. Bubonic plague: Yersinia pestis cc. Chancroid: Haemophilus ducreyi dd. Primary syphilis: T. pallidum ee. Gonococcal Infections ff. Syphilis gg. Treponematoses hh. Mycoplasma pneumoniae infections ii. Chlamydial infections jj. C. trachomatis infections kk. Meningitis, cerebritis and brain abscess ll. Bacterial meningitis mm. Brain abscess, suppurative intracranial infections nn. Cerebral malaria: Plasmodium falciparum
00. Spinal epidural abscess pp. Japanese encephalitis qq. Erysipelas rr. Cellulitis ss. Folliculitis tt. Myositis and myonecrosis uu. Tetanus vv. Legionella infections ww. Infective gastroenteritis xx. Pneumonia yy. Acute respiratory distress syndrome (ARDS) zz. Lung abscess aaa. Infective endocarditis
3. Fungal infections: Mycoses a. Coccidioidomycosis b. Histoplasmosis c. Blastomycosis d. Phaeohyphomycosis e. Penicilliosis f. Sporotrichosis g. Paracoccidioidomycosis h. Candidiasis i. Aspergillosis j. Cryptococcosis k. Mucormycosis (zygomycosis) I. Scedosporiosis m. Trichosporonosis n. Fusariosis
0. Pneumocystosis
4. Protozoal infections a. Entamoeba histolytica b. Malaria: Plasmodium falciparum c. Babesiosis d. Leishmaniasis e. Chagas disease and African trypanosomiasis f. Toxoplasmosis g. Trichomoniasis
5. Helmintic infections Cardiovascular diseases and neurovascular diseases
1. Brain a. Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) b. Ischaemic stroke c. Aneurysmal subarachnoid haemorrhage d. Cerebral ischaemia following subarachnoid haemorrhage e. Cerebral vasospasm
2. Atherosclerosis
3. Systemic arterial hypertension
4. Pulmonary hypertension
5. Deep venous thrombosis and pulmonary embolism
6. Heart a. Angina pectoris b. Myocardial ischaemia c. Myocardial infarction. d. Myocardial stunning e. Myocardial hibernation f. Post-ischaemic myocardial dysfunction g. Ischaemic cardiomyopathy h. Atrial and ventricular arrhythmia including atrial fibrillation i. Post-myocardial infarction syndrome j. Post-pericardiotomy syndrome k. Pericarditis Ll Myocarditis m. Rheumatic fever n. Cardiac complication after brain damage e Stress cardiomyopathy e Broken heart syndrome e Cardiac dysfunction and arrhythmia after subarachnoid haemorrhage e Cardiac dysfunction and arrhythmia after traumatic brain injury Neuroinflammatory and neurodegenerative diseases
1. Alzheimer's disease
2. Parkinson's disease
3. Huntington's disease
4. Extrapyramidal symptoms a. Acute dystonic reactions b. Oculogyric crisis c. Akathisia d. Pseudo-parkinsonism e. Tardive dyskinesia f. Sydenham's chorea
5. Acute disseminated encephalomyelitis (ADEM)
6. Hashimoto's encephalopathy
7. Bickerstaff's encephalitis
8. Anti-N-Methyl-D-Aspartate (Anti-NMDA) Receptor Encephalitis
9. Spinocerebellar ataxias
10. Susac's syndrome
11. Tolosa-Hunt syndrome
12. Meniere's disease
13. Multiple sclerosis
14. Idiopathic inflammatory demyelinating diseases
15. Transverse myelitis
16. Neuromyelitis optica {Devic’s disease)
17. Optic neuritis
18. Balo concentric sclerosis
19. Acquired neuropathies a. Chronic secondary polyneuropathies,
b. Chronic inflammatory demyelinating polyneuropathy (CIDP) c. Progressive inflammatory neuropathy d. Guillain Barré syndrome e. Critical illness polyneuropathy f. Acute and chronic motor axonal neuropathy
20. Hereditary neuropathies. a. Charcot-Marie-Tooth disease type 1, 2 and 3 b. Hereditary neuralgic amyotrophy
21. Myotonic dystrophy type | and II
22. Amyotrophic lateral sclerosis (ALS)
23. Neuralgic amyotrophy (Parsonage-Turner Syndrome)
24. Neuromyotonia
25. Myasthenia gravis
26. Restless legs syndrome
27. Stiff-person syndrome Epileptic disorders
1. Generalised epileptic seizures (grand mal) including status epilepticus
2. Focal epileptic seizures a. Frontal lobe epilepsy b. Temporal lobe epilepsy c. Benign Rolandic epilepsy d. Benign occipital epilepsy of childhood
3. Autosomal dominant nocturnal frontal lobe epilepsy
4. Childhood absence epilepsy
5. Dravet syndrome
6. Epilepsy in females with mental retardation
7. Juvenile myoclonic epilepsy
8. Lennox-Gastaut syndrome
9. Febrile infection-related epilepsy syndrome
10. West syndrome
11. Ohtahara syndrome
12. Reflex epilepsies
13. Progressive myoclonic epilepsies
14. Rasmussen's encephalitis Affective disorders and psychiatric syndromes
1. Schizophrenia
2. Depression
3. Bipolar disorder
4. Occupational burnout
5. Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcus (PANDAS)
6. Attention deficit hyperactivity disorder
7. Posttraumatic stress disorder
8. Fibromyalgia Fibrosis Primary fibrosis
1. Interstitial pulmonary fibrosis (ILD)
1. Retroperitoneal fibrosis
2. Primary biliary cholangitis (primary biliary cirrhosis or primary liver cirrhosis) Disease-induced fibrosis
1. Secondary pulmonary fibrosis
2. Secondary inflammation due to cystic fibrosis
3. Primary sclerosing cholangitis
4. Interstitial bladder fibrosis
5. Secondary liver cirrhosis a. Alcoholic cirrhosis b. Hepatitis C-induced cirrhosis c. Human immunodeficiency virus-induced cirrhosis d. Metastatic carcinomatosis cirrhosis Cancer-induced fibrosis Fibrosis formation induced by cancer
Tumour pseudoprogression Treatment induced fibrosis mimicking tumour progression especially in neuroendocrine tumours Treatment-induced fibrosis
1. Fibrosis formation induced by medical, surgical or radioactive treatments, i.e. Post abdominal operation peritoneal fibrosis with or without ileus
2. Fibrosis following organ transplantation
3. Chemotherapy-induced chronic gastrointestinal fibrosis Paraneoplastic syndromes
1. Paraneoplastic cerebellar degeneration
2. Lambert-Eaton myasthenic syndrome
3. Paraneoplastic cerebellar degeneration
4. Encephalomyelitis, limbic encephalitis
5. Brainstem encephalitis
6. Opsoclonus myoclonus ataxia syndrome
7. Anti-NMDA receptor encephalitis
8. Polymyositis
9. Acanthosis nigricans
10. Dermatomyositis
11. Leser-Trélat sign
12. Necrolytic migratory erythema
13. Sweet's syndrome
14. Florid cutaneous papillomatosis
15. Pyoderma gangrenosum
16. Acquired generalized hypertrichosis Cancer and neoplasms Immune-related and neoplastic blood diseases
1. Aplastic anaemia
2. Antiphospholipid syndrome (APS, APLS)
3. Thrombotic thrombocytopenic purpura (TTP)
4. ldiopathic thrombocytopenic purpura (ITP)
5. Autoimmune haemolytic anaemia
6. Autoimmune lymphoproliferative syndrome
7. Autoimmune neutropenia
8. Cold agglutinin disease
9. Essential mixed cryoglobulinemia
10. Evans syndrome
11. Pernicious anaemia
12. Pure red cell aplasia
13. Thrombocytopenia
14. Lymphangitis carcinomatosis
15. Myelodysplastic/myeloproliferative neoplasms (MPN/MPD) a. Polycythaemia vera b. Essential thrombocythemia c. Primary myelofibrosis d. Fibrotic myelofibrosis e. Lymphomas e Non-Hodgkin Lymphoma e Non-Hodgkin Lymphoma in Children (Burkitt lymphoma) e Hodgkin Disease. e Lymphoma of the skin e Waldenstrom macroglobulinemia + Multiple myeloma (Kahler’s disease) e Primary cerebral lymphoma f. Leukaemia e Acute myeloid leukaemia e Chronic myeloid leukaemia e Acute lymphocytic leukaemia e Chronic lymphocytic leukaemia g. Unclassifiable MPN/MPD Solid tumours
1. Neuroendocrine tumours (NETS) a. Pituitary gland: NET of the anterior pituitary b.
Thyroid gland: Neuroendocrine thyroid tumours and medullary carcinoma c.
Parathyroid NETS d.
Thymus and mediastinal carcinoid tumours e.
Pulmonary NETs e Bronchial NETS e Pulmonary carcinoid tumours: typical carcinoid and atypical carcinoid e Small-cell lung cancer (SCLC) e Large cell neuroendocrine carcinoma of the lung (LCNEC) e Extrapulmonary small cell carcinomas (ESCC or EPSCC) f.
Gastroenteropancreatic neuroendocrine tumours (GEP-NET) e Foregut NETs (stomach, proximal duodenum, thymus, lung and bronchus) e Pancreatic NETS e Midgut GEP-NET (from distal half of 2nd part of the duodenum to the proximal two-thirds of the transverse colon), appendix NETs e Hindgut NETS g.
Liver and gallbladder NETS h.
Adrenal tumours and adrenomedullary tumours i.
Pheochromocytoma j.
Peripheral nervous system NETS e Schwannoma e Paraganglioma e Neuroblastoma k.
Breast NETS I.
Genitourinary tract NETS e urinary tract carcinoid tumour and neuroendocrine carcinoma e ovary NETs e NETs of the cervix e Prostate NETS e Testes NETs m.
Merkel cell carcinoma of skin (trabecular cancer) n.
Hereditary NETS Multiple endocrine neoplasia type 1 (MEN1) and type 2 (MEN2) e Von Hippel-Lindau (VHL) disease e Neurofibromatosis type 1 and type 2 e Schwanomatosis e Tuberous sclerosis e Carney complex (LAMB or NAME syndrome)
2. Central nervous system tumours a. Brain tumours and spinal tumours e Metastatic brain tumours e Pilocytic astrocytoma e Glioma e Glioblastoma multiforme (GBM) e Oligodendroglioma e Ependymoma + Medulloblastoma e Meningeal tumours e Meningioma e Metastatic meningitis carcinomatosis
3. Oropharyngeal tumours a. Benign tumours e Eosinophilic granuloma e Fibroma e Granular cell tumour + Keratoacanthoma e Leiomyoma e Osteochondroma es Lipoma e Schwannoma e Neurofibroma e Papilloma e Condyloma acuminatum + Verruciform xanthoma e Pyogenic granuloma e Rhabdomyoma e Odontogenic tumours + Precancerous lesions e Leukoplakia e Erythroplakia e Erythroleukoplakia e Malignant tumours e Squamous cell carcinoma + Verrucous carcinoma e Minor salivary gland carcinomas e Lymphomas
4. Laryngeal cancer a. Paranasal sinus and nasal cavity cancer b. Nasopharynx cancer c. Teratomas d. Adenocarcinomas e. Adenoid cystic carcinomas f. Mucoepidermoid carcinomas. g. Salivary gland cancer h. Thyroid cancers + Papillary thyroid cancer Non-invasive follicular thyroid neoplasm with papillary-like nuclear features e Follicular thyroid cancer e Poorly differentiated thyroid cancer + Anaplastic thyroid cancer e Thyroid lymphoma e Squamous cell thyroid carcinoma e Sarcoma of thyroid + Hürthle cell carcinoma
5. Skin cancers a. Basal-cell carcinoma b. Squamous-cell carcinoma c. Malignant melanoma d. Kaposi sarcoma
6. Cancer of the vascular system a. Angiosarcomas b. Epithelioid haemangioendotheliomas c. Haemangiopericytomas d. Lymphangiosarcomas. e. Kaposiform haemangioendotheliomas (KHEs)
f. Infantile haemangioma g. Congenital haemangioma h. Haemangioblastoma i. Pyogenic granuloma j. Tufted angioma k. Glomus tumour
7. Muscle and bone tumours a. Leiomyoma b. Leiomyosarcoma c. Smooth muscle tumour of uncertain malignant potential (STUMP) d. Metastatic tumours e. Osteosarcoma f. Chondrosarcoma g. Ewing's sarcoma h. Fibrosarcoma i. Undifferentiated pleomorphic sarcoma j. Teratomas k. Osteoma I. Osteoid osteoma m. Osteochondroma n. Osteoblastoma o. Enchondroma p. Giant cell tumour of bone dq. Aneurysmal bone cyst
8. Breast cancer a. Metastatic tumours b. Invasive ductal carcinoma c. Invasive lobular carcinoma d. Tubular carcinoma e. Mucinous (colloid) carcinoma f. Carcinomas with medullary features g. Invasive papillary carcinoma h. Breast lymphoma i. Breast sarcoma
9. Lung tumours a. Metastatic tumours b. Bronchial leiomyoma c. Primary lung cancers ¢ Small-cell lung carcinoma (SCLC) e Non-small-cell lung carcinoma NSCLC) e Pleuropulmonary blastoma e Lymphomas of the lung e Sarcomas of the lung + Mediastinal tumours e Pleural tumours e Malignant mesothelioma e Pleural sarcomas e Pleural angiosarcoma e Pleural desmoplastic small round cell tumour (pleural DSRCT) e Pleural synovial sarcoma e Pleural solitary fibrous tumour (pleural SFT) e Smooth muscle tumours of the pleura e Pleural carcinomas e Pleural mucoepidermoid carcinoma + Pleural pseudomesotheliomatous adenocarcinoma e Pleural calcified fibrous pseudotumour
10. Gastrointestinal tumours a. Krukenberg tumour (metastatic tumour) b. Oesophageal cancer e Squamous-cell carcinoma (ESCC) e (Oesophageal adenocarcinoma (EAC) e Barrett's oesophagus e Gastric cancer e Gastric adenocarcinoma e Signet ring cell carcinoma e Gastric lymphoma e Extranodal marginal zone B-cell lymphomas (MALT lymphoma) c. Carcinoid e Duodenal adenocarcinoma e Appendix e Carcinoid d. Pseudomyxoma peritonei e Colorectal tumours e. Colorectal polyp: adenoma, hyperplastic, juvenile, sessile serrated adenoma, traditional serrated adenoma, Peutz-Jeghers syndrome f. Cronkhite-Canada syndrome g. Polyposis syndromes: Juvenile MUTYH-associated, familial adenomatous and serrated polyposis h. Adenocarcinoma i. Familial adenomatous polyposis j. Hereditary nonpolyposis colorectal cancer k. Anal tumour: Squamous cell carcinoma | Liver cancer m. Metastatic tumours n. Hepatocellular carcinoma
0. Hepatoblastoma p. Hepatocellular adenoma g. Cavernous haemangioma r. Focal nodular hyperplasia s. Nodular regenerative hyperplasia t. Gallbladder cancer u. Cholangiocarcinoma v. Klatskin tumour w. Gallbladder adenocarcinoma x. Pancreatic cancer y. Exocrine tumours e Adenocarcinoma e Pancreatic ductal adenocarcinoma z. Cystic neoplasms e Serous microcystic adenoma e Intraductal papillary mucinous neoplasm e Mucinous cystic neoplasm e Solid pseudopapillary neoplasm + Pancreablastoma aa. Endocrine PanNETs e MALT lymphomas e Peritoneal tumours bb. Metastatic peritonitis carcinomatosis
11. Urogenital cancers a. Renal tumours e Metastatic tumour e Renal cell carcinoma (RCC) o Clear cell RCC o Papillary RCC o Chromophobe RCC o Collecting duct RCC e Clear cell sarcoma e Mesoblastic nephroma e Wilm's tumour (nephroblastoma) + Renal oncocytoma e Cystic nephroma e Angiomyolipoma e Metanephric adenoma + Renal medullary fibroma b. Ureteral cancer e Transitional cell carcinoma e Ureteral neoplasm c. Bladder tumours + Metastatic tumours e Papillary transitional cell carcinoma e Non-papillary transitional cell carcinoma e Squamous cell carcinoma + Adenocarcinomas e Sarcomas e Small cell carcinomas d.
Ovarian tumours + Malignant ovarian cancer o Epithelial cancers o Germ cell cancers o Stromal cancers e Benign o Surface epithelial tumours o Stromal tumours o Germ cell tumours e.
Uterine tumours e Uterine fibroids or leiomyomas f.
Cervical tumours e Cervical cancer o Squamous cell carcinoma o Adenocarcinoma o Adenosquamous carcinoma o Small cell carcinoma o Neuroendocrine tumour o Glassy cell carcinoma o Villoglandular adenocarcinoma e Cervical intraepithelial neoplasia g.
Testicular tumours e Germ cell tumours o Intratubular germ cell neoplasia o Seminoma o Spermatocytic tumour o Embryonal carcinoma o Yolk sac tumour e Trophoblastic tumours o Choriocarcinoma o Monophasic choriocarcinoma o Placental site trophoblastic tumour o Cystic trophoblastic tumour e Teratomas o Dermoid cyst o Epidermoid cyst o Monodermal teratoma (Carcinoid) o Primitive neuroectodermal tumour (PNET) o Nephroblastoma-like tumour o Teratomic tumour with somatic-type malignancy e Sex cord - gonadal stromal tumours o Leydig cell tumour o Sertoli cell tumour o Lipid rich variant o Sclerosing variant o Large cell calcifying variant o Intratubular Sertoli cell neoplasia in Peutz-Jeghers syndrome o Granulosa cell tumour o Adult type o Juvenile type o Thecoma fibroma group o Thecoma o Fibroma o Incompletely differentiated tumours o Mixed types tumours e Mixed germ cell and sex cord/gonadal stromal tumours o Gonadoblastoma o Germ cell-sex cord/gonadal stromal tumour, unclassified e Miscellaneous tumours of the testis o Lymphomas v" Primary testicular diffuse large B-cell lymphoma v" Mantle cell lymphoma of the testes Extranodal marginal zone B cell lymphoma of the testes V Extranodal NK/T-cell lymphoma, nasal type of the testes “ Peripheral T-cell lymphoma of the testes v" Activin receptor-like kinase-1-negative anaplastic large cell lymphoma of the testes v' Paediatric-type follicular lymphoma of the testes o Carcinoid o Tumours of ovarian epithelial types v Serous tumour of borderline malignancy v Serous carcinoma Y Well differentiated endometrioid tumour ¥v" Mucinous cystadenoma v" Mucinous cystadenocarcinoma v" Brenner tumour o Nephroblastoma o Paraganglioma + Haematopoietic tumours e Tumours of collecting ducts and rete o Adenoma o Carcinoma e Tumours of the paratesticular structures o Adenomatous tumour o Malignant and benign mesothelioma o Adenocarcinoma of the epididymis o Papillary cystadenoma of the epididymis o Melanotic neuroectodermal tumour o Desmoplastic small round cell tumour e Mesenchymal tumours of the spermatic cord and testicular adnexae o Lipoma o Liposarcoma o Rhabdomyosarcoma o Aggressive angiomyxoma o Angiomyofibroblastoma-like tumour (see myxoma) o Fibromatosis o Fibroma o Solitary fibrous tumour o Others e Secondary tumours of the testis h.
Urethral cancer e Transitional cell carcinoma e Squamous-cell carcinoma e Adenocarcinoma + Melanoma e Prostate tumours Metastatic prostate tumour e Benign prostatic hyperplasia + Prostate cancer i. Penile tumours e Phimosis e Penile cancer
12. Retroperitoneal tumours a. Solid tumours e Metastatic tumour e Fibroma, fibrosarcoma, malignant fibrous histiocytoma e Lipoma, liposarcoma e Leiomyoma, leiomyosarcoma e Desmoid tumours e Ganglioneuroma, ganglioneuroblastoma e Schwannoma, neurofibroma e Extragonadal germ cell tumour e Lymphoma e Lymphadenopathy b. Cystic tumours e Cystic lymphangioma e Cystic teratoma e Cystadenoma, cystadenocarcinoma e Cystic mesothelioma e Epidermoid cyst e Tarlov cyst (perineural cyst) Trauma and posttraumatic syndromes
1. Traumatic subarachnoid haemorrhage
2. Head injury including severe traumatic head injury
3. Cerebral ischaemia following traumatic brain injury
4. Polytrauma condition
Inflammation after organ transplantation of lung, kidney, heart, liver, etc.
Acute immune reaction after organ transplantation Chronic organ tissue rejection reaction by the host Fibrosis following organ transplantation

Claims (32)

CONCLUSIESCONCLUSIONS 1. Een antagonist van een zoogdierlijke P2X7 receptor voor toepassing bij de behandeling van een hyperinflammatoir syndroom bij een zoogdierlijke patiënt, door primair lymfeklier gerichte toediening van de P2X7 receptor antagonist in de patiënt tot een concentratie in de beoogde lymfeklieren welke concentratie boven het maximaal tolereerbare plasmaniveau van de antagonist in het zoogdier is.An antagonist of a mammalian P2X7 receptor for use in the treatment of a hyperinflammatory syndrome in a mammalian patient, by primarily lymph node targeted administration of the P2X7 receptor antagonist in the patient to a concentration in the target lymph nodes which concentration is above the maximum tolerable plasma level of the antagonist in the mammal. 2. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 1, waarbij de antagonist wordt toegediend tot een concentratie in de beoogde lymfeklieren die overeenkomt met de ICx voor genoemde receptor, waarbij de ICx boven het maximaal tolereerbare plasmaniveau van de antagonist in het zoogdier ligt, waarbij x = 10, bij voorkeur = 20, meer bij voorkeur = 30, nog meer bij voorkeur = 40 en met de meeste voorkeur ongeveer 50 is.The mammalian P2X 1 receptor antagonist for use according to claim 1, wherein the antagonist is administered at a concentration in the target lymph nodes corresponding to the ICx for said receptor, wherein the ICx is above the maximum tolerable plasma level of the said receptor. antagonist is in the mammal, where x = 10, preferably = 20, more preferably = 30, even more preferably = 40 and most preferably about 50. 3. De antagonist van een P2X;7-receptor van een zoogdier voor toepassing volgens conclusie 1 of 2, waarbij de lymfeklier gerichte toediening wordt gekozen uit topicale en invasieve toediening.The mammalian P2X;7 receptor antagonist for use according to claim 1 or 2, wherein the lymph node targeted administration is selected from topical and invasive administration. 4. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens conclusie 3, waarin de toediening topicaal is, gekozen uit transmucosale en transdermale toediening.The mammalian P2X7 receptor antagonist for use according to claim 3, wherein the administration is topical selected from transmucosal and transdermal administration. 5. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 4, waarin de antagonist lipofiel is.The mammalian P2X1 receptor antagonist for use according to claim 4, wherein the antagonist is lipophilic. 6. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 5, waarin de lipofiele antagonist een vrije base is.The mammalian P2X1 receptor antagonist for use according to claim 5, wherein the lipophilic antagonist is a free base. 7. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 5 of 6, waarin de lymfeklier gerichte toediening transmucosale toediening in de mondholte omvat.The mammalian P2X1 receptor antagonist for use according to claim 5 or 6, wherein the lymph node targeted administration comprises transmucosal administration into the oral cavity. 8. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens conclusie 7, waarbij de toediening buccaal, sublinguaal, pharyngeaal of een combinatie daarvan is.The mammalian P2X7 receptor antagonist for use according to claim 7, wherein the administration is buccal, sublingual, pharyngeal or a combination thereof. 9. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens willekeurig welke van de conclusies 4 - 8, waarbij de toediening transdermaal is en in de vorm van een crème, zalf of lotion, patch of gips en/of waarbij micronaalden betrokken zijn of een combinatie daarvan.The mammalian P2X1 receptor antagonist for use according to any one of claims 4 to 8, wherein the administration is transdermal and in the form of a cream, ointment or lotion, patch or plaster and/or wherein microneedles involved or a combination thereof. 10. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 3, waarbij de toediening invasief is, gekozen uit intradermale, subdermale of subcutane toediening.The mammalian P2X1 receptor antagonist for use according to claim 3, wherein the administration is invasive selected from intradermal, subdermal or subcutaneous administration. 11. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 10, waarbij de antagonist hydrofiel is, in het bijzonder in de vorm van een in water oplosbaar farmaceutisch aanvaardbaar zout daarvan.The mammalian P2X1 receptor antagonist for use according to claim 10, wherein the antagonist is hydrophilic, particularly in the form of a water-soluble pharmaceutically acceptable salt thereof. 12. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 3, waarbij de toediening intraveneus is en de antagonist lipofiel is en wordt opgesloten in een geneesmiddel-afgiftesysteem, waarbij directe afgifte in het bloed wordt voorkomen.The mammalian P2X1 receptor antagonist for use according to claim 3, wherein the administration is intravenous and the antagonist is lipophilic and is entrapped in a drug delivery system, thereby preventing immediate delivery into the blood. 13. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij de P2X7-receptor activatie wordt geactiveerd door extracellulair ATP.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the P2X7 receptor activation is activated by extracellular ATP. 14. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarin de toediening een onmiddellijke afgiftedosering of een aanhoudende afgiftedosering is.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the administration is an immediate release dose or a sustained release dose. 15. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij de toediening een of meer bolustoedieningen continue toediening of een combinatie daarvan omvat.The mammalian P2X1 receptor antagonist for use according to any preceding claim, wherein the administration comprises one or more bolus administrations of continuous administration or a combination thereof. 16. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 15, waarbij een bolusdosering overeenkomt met ten minste 1.000 maal de hoeveelheid van de receptorantagonist, die aanwezig is in 1 ml plasma bij het maximaal tolereerbare plasmaniveau van de antagonist, bij voorkeur met ten minste 5.000 maal, met meer voorkeur met ten minste 10.000 maal.The mammalian P2X1 receptor antagonist for use according to claim 15, wherein a bolus dose corresponds to at least 1,000 times the amount of the receptor antagonist present in 1 ml of plasma at the maximum tolerable plasma level of the antagonist. preferably at least 5,000 fold, more preferably at least 10,000 fold. 17. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 15 of 18, waarbij de bolus 2 - 10 keer per dag wordt toegediend.The mammalian P2X1 receptor antagonist for use according to claim 15 or 18, wherein the bolus is administered 2-10 times per day. 18. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij de antagonist wordt toegediend in een vloeibaar medium dat ten minste 1 w/v % van de receptorantagonist bevat, bij voorkeur ten minste 5 w/v% en bij voorkeur ten minste 10 w/v%.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the antagonist is administered in a liquid medium containing at least 1 w/v% of the receptor antagonist, preferably at least 5 wt. /v% and preferably at least 10 w/v%. 19. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusies 10 en 15, waarbij de lymfeklier gerichte toediening plaatsvindt door middel van een continue intradermale, subcutane of subcutane infusie.The mammalian P2X1 receptor antagonist for use according to claims 10 and 15, wherein the lymph node targeted administration is by continuous intradermal, subcutaneous or subcutaneous infusion. 20. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens conclusie 19, waarin de dosering overeenkomt met ten minste 10 keer de IC:waarde per kg lichaamsgewicht per uur.The mammalian P2X7 receptor antagonist for use according to claim 19, wherein the dosage corresponds to at least 10 times the IC: value per kg of body weight per hour. 21. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarin de behandeling een hyperinflammatoir syndroom betreft van een ziekte, die is gekozen uit de groep, die bestaat uit auto- immuunziekten en immuun-gerelateerde ziekten zoals astma, allergie en chronische longziekten; behandelingsgeïnduceerde immuungerelateerde ziekten, zoals chemotherapie; infectieziekten, zoals virus- en bacteriële infecties; hart- en vaatziekten en neurovasculaire ziekten; neuro-inflammatoire en neurodegeneratieve ziekten; epileptische aandoeningen; affectieve stoornissen en psychiatrische syndromen; fibrose; kankergerelateerde aandoeningen; pseudoprogressie van tumoren; kanker en neoplasma's; trauma- en posttraumatische syndromen; post-orgaantransplantatie- syndromen met inbegrip van afstoting van getransplanteerde organen.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the treatment involves a hyperinflammatory syndrome of a disease selected from the group consisting of autoimmune diseases and immune-related diseases such as asthma, allergy and chronic lung diseases; treatment-induced immune-related diseases, such as chemotherapy; infectious diseases, such as viral and bacterial infections; cardiovascular and neurovascular diseases; neuroinflammatory and neurodegenerative diseases; seizure disorders; affective disorders and psychiatric syndromes; fibrosis; cancer-related conditions; pseudoprogression of tumors; cancer and neoplasms; trauma and post-traumatic syndromes; post-organ transplant syndromes including organ transplant rejection. 22. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij het hyperinflammatoire syndroom dyspneu omvat.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the hyperinflammatory syndrome comprises dyspnea. 23. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 22, waarin de dyspneu geassocieerd is met een virale infectie, bacteriële infectie, carcinomen, chronische obstructieve longziekte (COPD), astma, allergie, chemotherapie.The mammalian P2X1 receptor antagonist for use according to claim 22, wherein the dyspnea is associated with a viral infection, bacterial infection, carcinomas, chronic obstructive pulmonary disease (COPD), asthma, allergy, chemotherapy. 24. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de conclusies 21 - 23, waarbij de virale infectie wordt veroorzaakt door een virus, gekozen uit de groep, die bestaat uit Corona, in het bijzonder SARS-CoV-2; Influenza; Ebola; Respiratoir Syncytieel Virus; HIV.The mammalian P2X7 receptor antagonist for use according to any one of claims 21 to 23, wherein the viral infection is caused by a virus selected from the group consisting of Corona, especially SARS-CoV -2; Influenza; Ebola; Respiratory Syncytial Virus; HIV. 25. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij de P2X;-receptorantagonist gekozen is uit de groep, die bestaat uit: aminozuurderivaten, met name lidocaine, bupivacaine, ropivacaine en mepivacaine; antilichamen tegen P2x7zreceptoren, met name monoklonale antilichamen, aminozuurderivaten, met name benzocaïne en procaine; adamantane amidederivaten; triazoolderivaten; diarylimidazolidinederivaten; pyroglutaminezuur-The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the P2X1 receptor antagonist is selected from the group consisting of: amino acid derivatives, in particular lidocaine, bupivacaine, ropivacaine and mepivacaine; antibodies to P2x7 receptors, especially monoclonal antibodies, amino acid derivatives, especially benzocaine and procaine; adamantane amide derivatives; triazole derivatives; diarylimidazolidine derivatives; pyroglutamic acid amidederivaten; pyrazolacetamidederivaten; dihydrodibenzo [a,g] chinoliziniumderivaten; tetrazolderivaten; op tyrosine gebaseerde derivaten; pyrazooldiazepinederivaten; imidazoolderivaten; benzamidederivaten, KN62-analogen en -derivaten; adamantane carboxamiden; arylcarbohydraziden; cyaanguanidinen; aryltetrazolen en aryltriazolen; PPADS-tetranatriumzout; briljantblauw G (BBG); geoxideerd ATP (o-ATP); massadine; stylissadine A en B; P2X7-receptorremmers C23, C40 en C60; [3H]A-804598 ([3H]2-cyaan- 1-[(1S)-1-fenylethyl]-3-chinoline-5-yl-guanidine); bicycloheteroarylverbindingen.amide derivatives; pyrazolacetamide derivatives; dihydrodibenzo [a,g] quinolizinium derivatives; tetrazole derivatives; tyrosine-based derivatives; pyrazoldiazepine derivatives; imidazole derivatives; benzamide derivatives, KN62 analogs and derivatives; adamantane carboxamides; arylcarbohydrazides; cyanoguanidines; aryltetrazoles and aryltriazoles; PPADS tetrasodium salt; brilliant blue G (BBG); oxidized ATP (o-ATP); mass dinner; stylissadine A and B; P2X7 receptor inhibitors C23, C40 and C60; [3H]A-804598 ([3H]2-cyano-1-[(1S)-1-phenylethyl]-3-quinolin-5-yl-guanidine); bicycloheteroaryl compounds. 26. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens willekeurig welke van de voorgaande conclusies, waarbij de P2X7-receptorantagonist lidocaine bevat.The mammalian P2X7 receptor antagonist for use according to any preceding claim, wherein the P2X7 receptor antagonist contains lidocaine. 27. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens conclusie 26, waarbij de behandeling bestaat uit lokale toediening van lidocaine in de vrije base vorm.The mammalian P2X7 receptor antagonist for use according to claim 26, wherein the treatment consists of local administration of lidocaine in the free base form. 28. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 27, waarbij de behandeling de toediening van lidocaïne in de mondholte omvat.The mammalian P2X1 receptor antagonist for use according to claim 27, wherein the treatment comprises the administration of lidocaine into the oral cavity. 29. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 28, waarbij de lidocaïnebase wordt toegediend in een vloeibaar medium dat ten minste 2,5 w/v % van de receptorantagonist bevat, bij voorkeur ten minste 5 w/v %, en bij voorkeur ten minste 10 w/v %.The mammalian P2X i receptor antagonist for use according to claim 28, wherein the lidocaine base is administered in a liquid medium containing at least 2.5 w/v% of the receptor antagonist, preferably at least 5 w/v. v%, and preferably at least 10 w/v%. 30. De antagonist van een P2X;7-receptor van een zoogdier voor toepassing volgens conclusie 26, waarbij de behandeling invasieve toediening van lidocaïne in een in water oplosbare zoutvorm, met name lidocaïne-HCI omvat.The mammalian P2X;7 receptor antagonist for use according to claim 26, wherein the treatment comprises invasive administration of lidocaine in a water-soluble salt form, especially lidocaine HCl. 31. De antagonist van een P2X7-receptor van een zoogdier voor toepassing volgens conclusie 30, waarbij het lidocainezout intradermaal, subdermaal of subcutaan wordt toegediend.The mammalian P2X7 receptor antagonist for use according to claim 30, wherein the lidocaine salt is administered intradermally, subdermally or subcutaneously. 32. De antagonist van een P2X;-receptor van een zoogdier voor toepassing volgens conclusie 31, waarbij het lidocainezout wordt toegediend door middel van een continue intradermale, subcutane of subcutane infusie.The mammalian P2X1 receptor antagonist for use according to claim 31, wherein the lidocaine salt is administered by continuous intradermal, subcutaneous or subcutaneous infusion.
NL2027321A 2020-03-31 2021-01-14 Treatment of hyperinflammatory syndrome NL2027321B1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
NL2027321A NL2027321B1 (en) 2021-01-14 2021-01-14 Treatment of hyperinflammatory syndrome
JP2022559877A JP2023521623A (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
CA3178140A CA3178140A1 (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
EP21717576.9A EP4125843A1 (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
IL296719A IL296719A (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
AU2021248263A AU2021248263B2 (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
US17/907,640 US20230128348A1 (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
BR112022019824A BR112022019824A2 (en) 2020-03-31 2021-03-31 HYPERINFLAMMATORY SYNDROME TREATMENT
PCT/NL2021/050208 WO2021201680A1 (en) 2020-03-31 2021-03-31 Treatment of hyperinflammatory syndrome
CN202180025176.0A CN115605194A (en) 2020-03-31 2021-03-31 Treatment of excessive inflammatory syndrome

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
NL2027321A NL2027321B1 (en) 2021-01-14 2021-01-14 Treatment of hyperinflammatory syndrome

Publications (1)

Publication Number Publication Date
NL2027321B1 true NL2027321B1 (en) 2022-07-25

Family

ID=82557160

Family Applications (1)

Application Number Title Priority Date Filing Date
NL2027321A NL2027321B1 (en) 2020-03-31 2021-01-14 Treatment of hyperinflammatory syndrome

Country Status (1)

Country Link
NL (1) NL2027321B1 (en)

Similar Documents

Publication Publication Date Title
AU2021248263B2 (en) Treatment of hyperinflammatory syndrome
Wong et al. N-acetylcysteine as adjuvant therapy for COVID-19–a perspective on the current state of the evidence
JP5046922B2 (en) A therapeutic composition comprising at least one pyrrolobenzodiazepine derivative and fludarabine
Cocelli et al. Comparison of effects of low-flow sevoflurane and desflurane anesthesia on neutrophil and T-cell populations
JP2013535437A5 (en)
JP2011511072A5 (en)
CN103002877A (en) A sustained release formulation of methotrexate as a disease-modifying antirheumatic drug (DMARD) and an anti-cancer agent
CN105164143A (en) Cholestosome vesicles for incorporation of molecules into chylomicrons
JP2017226691A (en) Novel formulations of volatile anesthetics and methods of use thereof for reducing inflammation
US20190262267A1 (en) Pharmaceutical solution having a toxicity-reducing effect for antitumor drugs, and pharmaceutical composition comprising same
JP2005008534A (en) Anticancer agent and method for treating cancer
TW200816994A (en) Compositions and methods for the treatment of mucormycosis and other fungal diseases
JP2011518168A5 (en)
NL2027321B1 (en) Treatment of hyperinflammatory syndrome
TW202237151A (en) Treatment of hemophilia with fitusiran
Curigliano et al. Life-threatening toxic epidermal necrolysis during voriconazole therapy for invasive aspergillosis after chemotherapy
Zappavigna et al. Stealth liposomes for the delivery of zoledronic acid into tumors enhance the anticancer activity of the drug
JP2021522205A (en) Treatment of sepsis and septic shock
US11304947B2 (en) Compositions and methods for treating lung injuries associated with SARS-COV-2 infections
Kaur et al. Nascent Nanoformulations as an Insight into the Limitations of the Conventional Systemic Antifungal Therapies
US20230226211A1 (en) Cd25-targeted il-2 for increasing cd4 t cell formation and treatment of infections
US20080293804A1 (en) Pharmaceutical Nasal Spray Formulation of Acetaminophen (paracetamol)
Patil et al. Insight of Engineered Nano-based Biologics Approaches used to Combat Autoimmune Disease using TNF-α & IL Inhibitors
Jones COPD-Pathology, Diagnosis, Treatment, and Future Directions: Pathology, Diagnosis, Treatment, and Future Directions
GB2607584A (en) Composition and method of treatment

Legal Events

Date Code Title Description
PD Change of ownership

Owner name: NOMOREITIS B.V.; BE

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: DJOHAN HASAN

Effective date: 20220916