[go: up one dir, main page]

WO2025083710A1 - An antiviral photosensitive composition and device for potentiating the same - Google Patents

An antiviral photosensitive composition and device for potentiating the same Download PDF

Info

Publication number
WO2025083710A1
WO2025083710A1 PCT/IN2024/052076 IN2024052076W WO2025083710A1 WO 2025083710 A1 WO2025083710 A1 WO 2025083710A1 IN 2024052076 W IN2024052076 W IN 2024052076W WO 2025083710 A1 WO2025083710 A1 WO 2025083710A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
peg
acid
leds
antiviral
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/IN2024/052076
Other languages
French (fr)
Inventor
Vijay Kumar Ranka
Komal MINHAS
Dr. Vinayaka -
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Focelite Goodness India Private Ltd
Original Assignee
Focelite Goodness India Private Ltd
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
Application filed by Focelite Goodness India Private Ltd filed Critical Focelite Goodness India Private Ltd
Publication of WO2025083710A1 publication Critical patent/WO2025083710A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/062Photodynamic therapy, i.e. excitation of an agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/0613Apparatus adapted for a specific treatment
    • A61N5/0624Apparatus adapted for a specific treatment for eliminating microbes, germs, bacteria on or in the body
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N2005/065Light sources therefor
    • A61N2005/0651Diodes

Definitions

  • the present invention relates to an antiviral photosensitive composition comprising a phenothiazinium compound, and more particularly to a device for potentiating the phenothiazinium compound for treatment of dengue virus (DENV) infections.
  • DEV dengue virus
  • Dengue is an acute febrile illness resulting from the infection caused by four serologically related positive-strand ribonucleic acid (RNA) viruses belonging to the Flavivirus genus, namely dengue viruses (DENV) 1, 2, 3, and 4. Annually, an estimated 390 million dengue infections occur globally, resulting in up to 36,000 fatalities. [003] It is primarily a mosquito-borne viral infection that can lead to a wide range of clinical manifestations and severity. DENV infection is generally characterized by high fever, retro orbital pain, myalgia, leukopenia, thrombocytopenia, and hemorrhagic manifestations. Majority of these cases recover within 4–7 days without requiring significant intervention.
  • RNA positive-strand ribonucleic acid
  • DENV infection is generally characterized by high fever, retro orbital pain, myalgia, leukopenia, thrombocytopenia, and hemorrhagic manifestations. Majority of these cases recover within 4–7 days without requiring significant intervention.
  • dengue can progress to life threatening severe forms, such as Dengue Hemorrhagic Fever (DHF) or Dengue Shock Syndrome (DSS). Severe dengue is characterized by plasma leakage, severe bleeding, or organ dysfunction. Further, severe infections result in a significant clinical and economic burden due to the necessity for hospital admissions.
  • DHF Dengue Hemorrhagic Fever
  • DSS Dengue Shock Syndrome
  • severe dengue is characterized by plasma leakage, severe bleeding, or organ dysfunction. Further, severe infections result in a significant clinical and economic burden due to the necessity for hospital admissions.
  • Antibody-dependent enhancement is a critical mechanism that can worsen the disease during secondary infections. In this process, a complex interaction between the virus and the host’s immune system intensifies the severity of the infection.
  • the principal object of embodiments herein is to disclose a device for potentiating at least one phenothiazinium compound in a user and method for operating the same, wherein the device is wearable on a wrist of the user and the process of potentiation is performed using a plurality of LEDs present on the underside of the device and in contact with venous side of the wrist of the user.
  • Another object of embodiments herein is to disclose an antiviral photosensitive composition.
  • Another object of embodiments herein is to disclose an antiviral photosensitive composition effective against RNA viruses.
  • Another object of embodiments herein is to disclose an antiviral photosensitive composition effective against dengue and serotypes thereof.
  • Another object of embodiments herein is to disclose a method for treating infections caused by RNA viruses in a user.
  • Another object of embodiments herein is to disclose a non-invasive treatment method for viral infections such as dengue, reducing the need for conventional antivirals.
  • FIG.1 depicts a device for potentiating methylene blue, according to embodiments as disclosed herein;
  • FIG.2 depicts the components of the device, according to embodiments as disclosed herein;
  • FIG. 4 is a flowchart depicting the process of potentiating methylene blue using the device, according to embodiments as disclosed herein;
  • FIG.5 is a flow diagram depicting the in vitro assay with VERO cells for testing the antiviral photosensitive composition, according to embodiments as disclosed herein;
  • FIG.6 is a flow diagram depicting dilution preparation and plate layout for the in vitro assay, according to embodiments as disclosed herein;
  • FIGs.7A and 7B are graphs depicting the IC50 values of the different concentrations of the antiviral photosensitive composition against different serotypes of the dengue virus (DENV 1-4), wherein FIG.
  • FIGS. 8A and 8B are graphs depicting the percentage viability/toxicity of the Vero cells by MTT assay using the antiviral photosensitive composition, wherein FIG. 8A depicts the antiviral photosensitive composition (with light exposure), and FIG.
  • FIGs.9A and 9B are example cell morphology images from FRNT assay of Vero cells, wherein FIG.9A depicts example images of untreated cells (Cell and Virus control); and FIG.9B depicts example cells infected with DENV-2 and treated with different concentration of the antiviral photosensitive composition (with light exposure), according to embodiments as disclosed herein; and [0025] FIGs.10A and 10B are example cell morphology images from FRNT assay of Vero cells, wherein FIG.
  • FIG. 10A depicts example images of untreated cells (Cell and Virus control); and FIG.10B depicts example cells infected with DENV-2 and treated with different concentration of the antiviral photosensitive composition (without light exposure), according to embodiments as disclosed herein.
  • DETAILED DESCRIPTION [0026] The embodiments herein and the various features and advantageous details thereof are explained more fully with reference to the non-limiting embodiments that are illustrated in the accompanying drawings and detailed in the following description. Descriptions of well-known components and processing techniques are omitted so as not to unnecessarily obscure the embodiments herein. The examples used herein are intended merely to facilitate an understanding of ways in which the embodiments herein may be practiced and to further enable those of skill in the art to practice the embodiments herein.
  • Embodiments herein may be described and illustrated in terms of blocks which carry out a described function or functions. These blocks, which may be referred to herein as managers, units, modules, hardware components or the like, are physically implemented by analog and/or digital circuits such as logic gates, integrated circuits, microprocessors, microcontrollers, memory circuits, passive electronic components, active electronic components, optical components, hardwired circuits and the like, and may optionally be driven by a firmware.
  • the circuits may, for example, be embodied in one or more semiconductor chips, or on substrate supports such as printed circuit boards and the like.
  • circuits constituting a block may be implemented by dedicated hardware, or by a processor (e.g., one or more programmed microprocessors and associated circuitry), or by a combination of dedicated hardware to perform some functions of the block and a processor to perform other functions of the block.
  • a processor e.g., one or more programmed microprocessors and associated circuitry
  • Each block of the embodiments may be physically separated into two or more interacting and discrete blocks without departing from the scope of the disclosure.
  • the blocks of the embodiments may be physically combined into more complex blocks without departing from the scope of the disclosure.
  • Embodiments herein disclose a device 100 for potentiating at least one phenothiazinium compound and method for operating the same. Referring now to the drawings, and more particularly to FIGs. 1 through 10B, where similar reference characters denote corresponding features consistently throughout the figures, there are shown embodiments. [0033] Embodiments herein use the terms ‘activation’, ‘activate’, ‘potentiate’, and so on interchangeably to refer to the process of enhancing the effect of a drug or a compound thereby allowing faster recovery.
  • the device 100 potentiates the antiviral properties of at least one phenothiazinium compound in an antiviral photosensitive composition. In an embodiment, the device 100 potentiates the antiviral properties of methylene blue in the antiviral photosensitive composition.
  • the device 100 can be in the form of a wearable device. In an embodiment herein, a person/subject can wear the device 100 on their wrist, wherein a bottom surface of the device 100 is placed in proximity to the underside of the person’s wrist (i.e., the venous side of the wrist) using a suitable means (such as, but not limited to, a strap).
  • the device 100 comprises a body 101 which can be at least one of temporarily or permanently attached to a strap 102.
  • the strap 102 can be used to attach the device 100 to the wrist of a person securely, such that the underside of the device 100 is in contact with the underside of the wrist of the person (i.e., the venous side of the wrist of the person).
  • Embodiments herein use the terms ‘user’, ‘subject’, ‘person’, and so on interchangeably to refer to the user on whose wrist to which the device is currently attached.
  • the device 100 does not comprise a strap, wherein the device 100 can be handheld against the underside of the person’s wrist (i.e., the venous side of the wrist) for a pre-defined period of time.
  • the body 101 of the device 100 as depicted in FIG.2, comprises a controller 201, a timing module 202, a communication module 203, one or more user interfaces 204, a plurality of Light Emitting Diodes (LEDs) 205, at least one battery 206, and a battery charging module 207.
  • the communication module 203 can enable one or more authorized users (such as a person administering the test/treatment, the person who is undergoing the test/treatment, a person who is supervising the test/treatment, and so on) to interact with the device 100.
  • the communication module 203 can use one or more of a wired interface or a wireless interface to enable the one or more authorized users to interact with the device 100. Examples of the interface can be, but not limited to, Wi-Fi, a cellular network, Bluetooth, Bluetooth Low Energy (BLE), Near Field Communication (NFC), a Universal Serial Bus (USB) port, and so on.
  • the one or more user interfaces 204 can enable the one or more authorized users to access and/or control the device 100.
  • Examples of the one or more user interfaces 204 can be, but not limited to, one or more switches, toggles, a display, a touchscreen, a plurality of lights (such as LEDs (not the same as the LEDs 205)), a speaker, a microphone, a dial, and so on.
  • the plurality of Light Emitting Diodes (LEDs) 205 can be placed on the bottom surface of the device 100, such as the LEDs are in proximity to the venous side of the wrist of the person, when the person is wearing the device 100.
  • the plurality of LEDs can be domed LEDs.
  • the plurality of LEDs can be non-domed LEDs.
  • the LEDs can have varying wavelengths.
  • the wavelength of the LEDs can range from 610 – 670 nm.
  • the number of LEDs 205 can comprise of two – twelve LEDs, arranged on the bottom surface of the device 100.
  • the device 100 can comprise of six LEDs, wherein of the six LEDS, three LEDs can have a wavelength of 670nm (i.e., red light) with an intensity of 30000 – 50000Lux (+/ -5%), and the other three LEDs can have a wavelength of 610nm (i.e., amber light) with an intensity of 15000 – 25000Lux (+/ -5%).
  • MB methylene blue
  • PDT photodynamic therapy
  • Redox potential reflects the MB's ability to accept or donate electrons.
  • MB absorbs light it can transition from an oxidized (MB+) state to a reduced state (leucomethylene blue, LMB) and back, depending on the environment and the available energy.
  • MB+ oxidized
  • LMB reduced state
  • 610 nm light has slightly higher photon energy ( ⁇ 2.03 eV) as compared to 670 nm ( ⁇ 1.85 eV).
  • the LEDs 205 can be arranged in parallel with each other. In an embodiment herein, the LEDs 205 can be arranged in series with each other. In an embodiment herein, the LEDs 205 can be arranged in series and parallel with each other. In an example herein, the LEDs 205 can provide a light output of 1.2V – 2.4V.
  • FIGs.3A and 3B depict example layouts of the LEDs on the device 100.
  • the at least one battery 206 can provide power to the device 100, as required.
  • the at least one battery 206 can be a Lithium-ion (Li-ion) battery.
  • the at least one battery 206 can be charged by the battery charging module 207.
  • the battery charging module 207 can use a suitable charging port for charging the battery 206. Examples of the charging port can be, but not limited to, a USB port, a micro-USB port, a USB-C port, a custom port, and so on.
  • the battery 206 can be charged using wireless charging.
  • the battery charging module 207 can further a comprise a visual and/or an audio indicator (not shown), indicating the charging status (the device 100 is to be charged, the device 100 is charging, current battery level(s), the device 100 has completed charging, and so on).
  • the term ‘controller 201,' as used in the present disclosure, may refer to, for example, hardware including logic circuits; a hardware/software combination such as a processor executing software; or a combination thereof.
  • the processing circuitry more specifically may include, but is not limited to, a central processing unit (CPU), an arithmetic logic unit (ALU), a digital signal processor, a microcomputer, a field programmable gate array (FPGA), a System-on- Chip (SoC), a programmable logic unit, a microprocessor, application-specific integrated circuit (ASIC), etc.
  • the controller 201 may include at least one of, a single processer, a plurality of processors, multiple homogeneous or heterogeneous cores, multiple Central Processing Units (CPUs) of different kinds, microcontrollers, special media, and other accelerators.
  • an authorized person can initiate the device operation for a pre-defined period of time.
  • the pre-defined period of time can depend on the time required for a blood cell to complete one circulation through the subject (which depends on one or more physiological parameters of the user, such as, but not limited to, age of the user, physical condition of the user, health conditions of the user, blood flow rate of the user, and so on), wherein the pre- defined period of time can be equal to one – ten periods of time required for a blood cell to complete one circulation through the human body, and can be the time required for potentiating the at least one phenothiazinium compound.
  • the pre-defined period of time can vary from 1 – 60 minutes.
  • the user can press a first switch to configure the device 100 to operate for a first period of time, the user can press a second switch to configure the device 100 to operate for a second period of time, and so on.
  • the user can use a touchscreen display to configure the device 100 to operate for a first period of time, the user can use a touchscreen display to configure the device 100 to operate for a second period of time, and so on.
  • the controller 201 can provide instructions to one or more LEDs to turn on/off for the configured pre- defined period of time.
  • the controller 201 can provide instructions to all the LEDs 205 to light up for the pre-defined period of time. On receiving this intimation to initiate the device operation, in an embodiment herein, the controller 201 can provide instructions to a pre-defined set of LEDs 205 to light up for the pre-defined period of time standardized for adult & children respectively. The controller 201 can monitor the time of the device operation using the timing module 202. [0045] The LEDs 205 can turn ON and activate at least one phenothiazinium compound. In an embodiment herein, the at least one phenothiazinium compound can be methylene blue (MB).
  • MB methylene blue
  • Light converts MB into a reduced form leucomethylene blue, a non-ionic compound that has the capability to modulate the immune system.
  • the LEDs 205 can pass through skin at depths ranging from 3 to 4 mm.
  • the controller 201 can turn off the LEDs 205.
  • the controller 201 can provide an alert using a visual and/or an audio indicator (not shown) to the user that the device operation has been successfully completed.
  • the authorized person (who can be at least one of a person authorized to perform the action, and the user to whose wrist the device is attached) initiates the device operation for the pre-defined period of time.
  • the controller 201 provides instructions to the LEDs to turn ON for the configured pre-defined period of time.
  • the LEDs 205 turns ON.
  • the controller 201 turns off the LEDs 205.
  • Embodiments herein further disclose a method for treating infections caused by RNA viruses in a subject. The method comprising administering an antiviral photosensitive composition including at least one phenothiazinium compound, at least one pharmaceutically acceptable carrier and water; and potentiating the least one phenothiazinium compound using the device 100.
  • the at least one phenothiazinium compound can be methylene blue.
  • the method includes administering the antiviral photosensitive composition, and potentiating methylene blue using the device 100.
  • the term "user” or “subject”, as used interchangeably herein, refers to a member of an animal species of mammalian origin that may benefit from the administration of described antiviral photosensitive composition or method of the described invention.
  • the user is a human and animal.
  • the user is infected by RNA viruses.
  • the user is infected by viruses belonging to Flaviviridae family, including but not limited to, West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Feline Infectious Peritonitis, Zika virus, and serotypes thereof.
  • viruses belonging to Flaviviridae family including but not limited to, West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Feline Infectious Peritonitis, Zika virus, and serotypes thereof.
  • the virus is dengue virus and serotypes thereof.
  • Embodiments herein disclose an antiviral photosensitive composition.
  • the composition includes at least one phenothiazinium compound; at least one pharmaceutically acceptable carrier selected from a group consisting of lipids, stabilizers, lubricants, and water.
  • photosensitive composition refers to a mixture containing at least one compound capable of photoactivation, particularly when exposed to light, particularly visible light which potentiates the redox potential of the molecule to convert into ionic and nonionic form along with free oxygen liberation. It includes photosensitizing agents or photosensitizers, salts, or prodrug, or analogs, or derivatives thereof.
  • Non limiting examples of photosensitizers include, prophyrins; dyes such as phenothiazinium, phenodiazinium, phenooxaziniums; chlorophyll; chlorins; bacteriochlorins; phenothiaziniums; prophycenes; and other natural occurring photosensitizing agents, or mixtures thereof.
  • the photosensitizer is a phenothiazinium compound.
  • the composition according to embodiments herein includes at least one phenothiazinium compound.
  • phenothiazinium refers to a group of heterocyclic compounds having phototoxic efficiency against a broad range of microorganisms.
  • phenothiazinium compounds includes, Methylene blue (MB), 1-Octanol, 1,3- diphenylisobenzofuran (DPIBF), Rose Bengal (RB), new methylene blue (NMB), dimethyl methylene blue (DMMB), Azure A (AA), azure C (AC), azure B (AB), toluidine blue O (TBO), brilliant crystal blue (BCB), pyronin Y (PYY), neutral red (NR), derivatives and analogs thereof.
  • the phenothiazinium compound is methylene blue.
  • Methylene Blue is a well-known tricyclic phenothiazinium compound with redox-active properties making it suitable for treating infectious diseases. It is approved by the FDA and EMA for the treatment of methemoglobinemia and malaria.
  • MB can oscillate between oxidized (MB+) and reduced (MBH) states. This redox cycling mechanism involves the reduction of MB to its leucomethylene blue form, which is then re-oxidized back to MB, creating a continuous cycle of redox reactions.
  • the ionic form of MB is used to target the viral protease, while the nonionic form (leucomethylene blue) is involved in modulating cytokine storm & the redox cycling mechanism.
  • MB Upon illumination, MB absorbs visible light energy and becomes activated, causing lipid envelope disintegration and RNA degradation in viruses, including those with double envelopes. These disrupt the viral membrane and cause destruction of the nucleic acids, particularly at guanosine residues. The resulting nucleic acid modification can prevent viral replication and induce the inactivation of viruses.
  • methylene blue disrupts the activity of NS1, a key viral protein essential for DENV replication and immune evasion.
  • MB interacts with proteins such as G3BP1, G3BP2, DDX6, Caprin1, and USP10, which are crucial for stress granule formation during DENV infection. This prevents viral replication by limiting stress granule assembly. Furthermore, MB regulates the unfolded protein response (UPR), activated under ER stress during infection, helping maintain ER homeostasis and reducing viral replication. [0057] MB also interferes with histone deacetylase (HDAC) activity, preventing chromatin condensation and restoring normal host transcription. It influences host gene expression by interacting with long non-coding RNAs (lncRNAs) and genes that promote viral replication through epigenetic regulation.
  • HDAC histone deacetylase
  • MB inhibits Positive Transcription Elongation Factor- b (P-TEFb), which helps reverse the overexpression of IL-8 associated with severe dengue hemorrhagic fever (DHF).
  • P-TEFb Positive Transcription Elongation Factor- b
  • DHF severe dengue hemorrhagic fever
  • PDT photodynamic therapy
  • the phenothiazinium compound is present in an amount ranging from 0.001%w/w to 0.02%w/w including all the values in the range, for instance 0.002%w/w, 0.003%w/w, 0.004%w/w, 0.005%w/w, and so on.
  • methylene blue is present in an amount ranging from 0.001%w/w to 0.02%w/w.
  • the composition includes at least one pharmaceutically acceptable carrier.
  • Non-limiting examples of pharmaceutically acceptable carriers includes solvents, diluents, lipids (fats and oils), binders, disintegrants, lubricants, preservatives, emulsifying agents, surfactants, stabilizers, humectants, buffers, flavoring and sweetening agents, coloring agents, thickening Agents (viscosity enhancers), coating agents, and so on.
  • the composition includes lipids.
  • Non-limiting examples of lipids include phosphatidic acid, cholesterol, triglycerides such as caprylic/capric triglycerides, soybean oil, castor oil, olive oil; fatty acids such as linoleic acid, lauric acid, myristic acid, capric acid, arachidonic acid; fatty alcohols cetearyl alcohol, cetyl alcohol, oleyl alcohol, stearyl alcohol, lanolin alcohol; glycerides such as glyceryl monostearate, glyceryl distearate, glyceryl tricaprylate, glyceryl tristearate, glyceryl dibehenate; lecithin such as soy lecithin, egg lecithin; phospholipids such as lecithin, phosphatidylcholine (PC), non-hydrogenated phosphatidylcholine, phosphatidylglycerol (PG), phosphatidylserine (PS), Soya phosphat
  • the lipid is non-hydrogenated phosphatidylcholine.
  • non-hydrogenated phosphatidylcholine acts as a binder in the present composition.
  • the lipid is present in an amount ranging from 3 %w/v to 4.5 %w/v including all the values in the range, for instance 3.1 %w/v, 3.2 %w/v, 3.3 %w/v, 3.4 %w/v, and so on.
  • non-hydrogenated phosphatidylcholine is present in an amount ranging from 3 %w/v to 4.5 %w/v.
  • the composition includes stabilizer.
  • Non-limiting examples of stabilizers include antioxidants such as ascorbic acid, butylated hydroxytoluene, butylated hydroxyanisole, sodium metabisulfite, tocopherols; chelating agents such as ethylenediaminetetraacetic Acid (EDTA), disodium EDTA, citric acid; polyvinylpyrrolidone (PVP), hydroxypropyl Methylcellulose (HPMC), carboxymethylcellulose (CMC); amino acids such as arginine, histidine, glycine; sugars and polyols such as sucrose, sorbitol, mannitol, glycerin, and so on.
  • the stabilizer is glycerin.
  • the stabilizer is present in an amount ranging from 1 %w/v to 2.5 %w/v including all the values in the range, for instance 1.1 %w/v, 1.2 %w/v, 1.3 %w/v, 1.4 %w/v, and so on.
  • glycerin is present in an amount ranging from 1 %w/v to 2.5 %w/v.
  • the composition includes lubricants.
  • Non-limiting examples of lubricants include magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, polyethylene glycol (PEG) or derivatives thereof, sodium benzoate, liquid paraffin, zinc stearate, calcium stearate, and so on.
  • the lubricant is polyethylene glycol (PEG) or derivatives thereof. Addition of PEG or PEG derivatives enhances the immunocompatibility of the active ingredient as PEGylated active ingredient exhibits prolonged in vivo circulation time resulting in their high bioavailability.
  • Non-limiting examples of PEG and PEG derivatives include PEG 100, PEG 200, PEG 300, PEG 400, PEG 500, PEG 600, PEG 700, PEG 800, PEG 900, PEG 1000, PEG 1100, PEG 1200, PEG 1300, PEG 1400, PEG 1500, PEG 1600, PEG 1700, PEG 1800, PEG 1900, PEG 2000, PEG 2100, PEG 2200, PEG 2300, PEG 2400, PEG 2500, PEG 2600, PEG 2700, PEG 152800, PEG 2900, PEG 3000, PEG 3250, PEG 3350, PEG 3500, PEG 3750, PEG 4000, PEG 4250, PEG 4500, PEG 4750, PEG 5000, PEG 5500, PEG 6000, PEG 6500, PEG 7000, PEG 7500, PEG 8000, PEG 8500, PEG 9000, PEG 9500, PEG 10,000, PEG 11,000, PEG 12,000,
  • the PEG is PEG 400.
  • the lubricant is PEG 400.
  • the lubricant is present in an amount ranging from 0.5 %w/v to 3 %w/v including all the values in the range, for instance 0.6 %w/v, 0.7 %w/v, 0.8 %w/v, 0.9 %w/v, and so on.
  • PEG 400 is present in an amount ranging from 0.5 %w/v to 3 %w/v.
  • the composition further includes water. The water is present in an amount sufficient to make 100%, i.e., water is in a quantity sufficient to make 100% of the composition.
  • the composition has a pH ranging from 5 to 6.
  • Table 1 provides the antiviral photosensitive composition, according to embodiments herein. S. No. Ingredient Qty. 1 Phenothiazinium compound 0.001 to 0.02 %w/w 4 Lipid 3 to 4.5 %w/v 5 Stabilizer 1 to 2.5 %w/v 6 Lubricant 0.5 to 3 %w/v 7 Purified water, USP QS to 100% [0069] It is understood that various such modifications in the composition would be apparent to a person skilled in the art in light of the disclosures made herein and are included within the scope of the embodiments herein. [0070] Embodiment herein further disclose a method for preparing the antiviral photosensitive composition.
  • the method includes encapsulating the at least one phenothiazinium compound in liposomes.
  • Various methods for preparing liposomes are generally known in the art which may be used in achieving the embodiments herein. These include, thin film hydration, ether/ethanol injection method, reverse phase evaporation method, detergent depletion method, heating method, microfluidic channel method, membrane extrusion method, homogenization and sonication method.
  • liposomes are prepared by homogenization and sonication method.
  • the method for preparing the antiviral photosensitive composition includes mixing at least one stabilizer in water to obtain a clear solution; dissolving at least one phenothiazinium compound in at least one solvent to obtain a phenothiazinium solution; mixing the clear solution, the phenothiazinium solution and adding at least one lipid; and sonicating and adding at least one lubricant to the resulting solution.
  • the method includes mixing at least one stabilizer in water to obtain a clear solution.
  • Mixing may be achieved by any suitable method generally known in the field including, but not limited to, blending, sonicating, magnetic stirring, etc., by using equipment such as sonicator, magnetic stirrer, blender, mixer, etc.
  • mixing is performed for a period ranging from 5 to 15 minutes.
  • mixing is performed at 1300 to 1700 rpm.
  • the at least one stabilizer is mixed in water for a period ranging from 5 to 15 minutes at 1300 to 1700 rpm to obtain a clear solution.
  • the method includes dissolving at least one phenothiazinium compound in at least one solvent to obtain a phenothiazinium solution.
  • Non-limiting examples of solvents include water, ethanol, chloroform, glacial acetic acid, glycerol, dimethyl formamide, and dimethyl sulfoxide.
  • the solvent is water.
  • methylene blue is dissolved in water.
  • the method further includes mixing the clear solution, the phenothiazinium solution and adding at least one lipid.
  • the lipid is non hydrogenated phosphatidylcholine.
  • the method further includes sonicating and adding at least one lubricant to the resulting solution. In an embodiment, sufficient quantity of water is added to make 100% of the composition before sonication. Sonication may be achieved by methods and conditions generally known in the art.
  • ultra-sonication is performed by passing the solution through a sonicator tunnel at a flow rate of 200 mL per minute, at 350 to 450 ultrasonic power. In an embodiment, sonication is performed for a time period ranging from 5 to 15 minutes.
  • the lubricant is polyethylene glycol.
  • the composition may be filtered using methods generally known in the art. In an embodiment, the composition is filtered through 0.2 ⁇ m acetate cellulose membranes. [0077] The composition, according to embodiments herein, may further be formulated into any suitable dosage form.
  • Non-limiting examples of dosage forms include, colloids, injectables, solution for oral or intravenous (IV)/ intramuscular (IM) administration, emulsions, suspensions, dispersion, capsules, tablets, lozenges, powder, lyophilized cakes, granules, gel, cream, lotions, and aerosols.
  • the dosage form of the composition is capsule.
  • the dosage form of the composition is a tablet.
  • Generally known methods of formulating/processing compositions may be used to formulate the desired dosage forms.
  • the disclosed composition is formulated in the form of capsule.
  • the disclosed composition is formulated in the form of tablets.
  • the composition can be administered through various routes, including, but not limited to, oral, intravenous (IV), intramuscular (IM), subcutaneous (SC), topical, inhalation, and transdermal.
  • the dosage form is suited for oromucosal route of administration e.g.: buccal, sublingual, etc.
  • the composition is administered intraorally, and the components be prepared for uptake in a manner that makes the composition available in therapeutically effective amounts. As such, they may also be prepared as water soluble compositions, deliverable in encapsulated, or in a manner suitable for time release, delayed release or any manner typically used for delivery of pharmaceuticals, nutraceuticals or vitamins, etc.
  • the dosage of the composition that may be used in the present invention will vary with the route of administration, the rate of excretion, the duration of the treatment, the identity of any other therapeutic compounds being administered, the age, size, and species of the subject, e.g., human patient, and like factors.
  • the dosage of a compound that may be used in the present invention will be an amount which is the lowest dose effective to produce the desired effect with minimal side effects or with very low side effect in comparison with therapeutic benefit.
  • the composition is administered in a dosage ranging from 1 to 15 mg per day which can be administered in single or multiple doses.
  • the device 100 combined with the antiviral photosensitive composition is effective against DENV and other RNA viruses.
  • Non-limiting examples of RNA viruses includes, Coronavirus family (Coronaviridae), Flavivirus family (Flaviviridae) such as West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Zika virus, and serotypes thereof; Orthomyxovirus family (Orthomyxoviridae) such as Influenza A, B, and C viruses; Retrovirus family (Retroviridae); Filovirus family (Filoviridae); Paramyxovirus family (Paramyxoviridae); Togavirus family (Togaviridae); and Picornavirus family (Picornaviridae).
  • the device 100 combined with the antiviral photosensitive composition are effective against dengue virus and serotypes thereof.
  • the dengue virus is DENV-1.
  • the dengue virus is DENV-2.
  • the dengue virus is DENV-3.
  • the dengue virus is DENV- 4.
  • the inventors of the present invention have achieved a methylene blue-based composition-device combination therapy where the redox potential of MB is exploited using photodynamic therapy (PDT). PDT further enhances MB’s effect by potentiating redox potential converting into ionic and nonionic form, maximizing viral inactivation.
  • PDT photodynamic therapy
  • the device 100 potentiates MB at 610-670 nm, generating redox cycle amplification of MB at the application site. MB cycles between oxidized and reduced states to modulate cellular oxidative stress, disrupting viral replication.
  • the methylene blue-based composition-device combination therapy provides an enhanced antiviral activity through combined form (ionic and non-ionic form) of molecule.
  • the therapy also targets the host body by reducing oxidative stress and inflammation, which are common complications of dengue virus infections.
  • the device 100 further provides precise wavelength activation ensuring efficient MB activation with minimal phototoxicity.
  • the inventors of the present invention further achieved an effective composition against dengue virus with even a low concentration (i.e.,1 to 2 mg) of methylene blue, thereby minimizing side effects of methylene blue.
  • the disclosed composition is capable of bypassing first pass metabolism.
  • the composition can overcome potential loss or reduction in active ingredient i.e., methylene blue caused due to first pass effect or pre-systemic metabolism, thereby improving its bioavailability and stability.
  • the present invention contributes to antimicrobial resistance (AMR) mitigation by reducing dependence on traditional antivirals.
  • AMR antimicrobial resistance
  • Example 1 Preparation of the antiviral photosensitive composition
  • 12,500 mg glycerin USP or BP Grade
  • the mixing was carried out at 1500 rpm for 10 minutes under room temperature to obtain a clear solution.
  • a solution of methylene blue was prepared by dissolving 10000mg methylene blue in 100ml ml of purified water. Both the glycerin solution and the methylene blue solution were mixed along with addition of 15000mg of non-hydrogenated phosphatidylcholine solution. Purified water was added to the resulting solution to bring the total volume to 900 mL.
  • the solution was sonicated using ultra sonicate probe by passing the solution through a sonicator tunnel at 200ml /min with 400UP (ultrasonic power) for 10 minutes. Subsequently, 1g/100ml of polyethylene glycol was added to the solution. The final volume was adjusted, and the composition was finally filtered through 0.2 ⁇ m acetate cellulose membranes.
  • the inventors of the present invention conducted a series of experiments to determine the optimal concentration of the phenothiazinium compound (methylene blue) in the antiviral photosensitive composition (along with light exposure) for maximum efficacy against different serotypes of the dengue virus.
  • Vero cells were used.
  • the Vero cell line derived from the kidney epithelial cells of the African Green Monkey, is highly susceptible to a wide range of viral infections, including many human and animal viruses. This characteristic makes them a versatile tool for studying and screening antiviral compounds against various types of viruses, including Dengue Virus (DENV). In addition to this, Vero cells do not produce interferon which help researchers to study effectiveness of antiviral compound.
  • DENV Dengue Virus
  • Vero cells do not produce interferon which help researchers to study effectiveness of antiviral compound.
  • 2.5 x 105 Vero cells were seeded per well in a 96-well plate with 100 ⁇ l of complete DMEM medium containing 5% ⁇ FBS (Fetal bovine serum).
  • the cells were then incubated in a humidified incubator at 37°C with 5% CO2 for 24 hours to allow them to adhere and attain proper morphology.
  • MOI Multiplicity of Infection
  • the infection was carried out in 40 ⁇ l of infection media per well, consisting of 1x DMEM with 2% ⁇ FBS, and the cells were incubated for 2 hours in a humidified incubator at 37°C with 5% CO2. After 2 hrs, the virus containing media was aspirated from the wells, followed by washing the well with 100 ⁇ l incomplete (1x DMEM) media.
  • FIG. 5 is a flow diagram depicting the in vitro assay using VERO cells to test the antiviral photosensitive composition.
  • the composition was further diluted by dissolving in media (DMEM + 2% FBS) to obtain a concentration of 100 ug/mL.
  • DMEM + 2% FBS dissolving in media
  • the plate was incubated for 10 minutes at RT to allow fixation. Followinged by centrifugation at 2500 rpm for 5 minutes at RT. The pellet was further washed twice with 150 ⁇ l/well of 1X Permeabilization Buffer (perm buffer). After every wash, centrifugation was carried out at 2500rpm for 5 minutes at RT. The cells were then treated with 40 ⁇ l/ well Blocking solution (1% Normal Mouse Serum in 1x Perm Buffer) followed by re-suspending the cells and incubating the plate at RT for 30 mins.20 ⁇ l/ well Alexa Fluor 488 labelled anti-DENV monoclonal antibody was then added along with blocking solution.
  • 1X Permeabilization Buffer perm buffer
  • the cells were then mixed and incubated in a plate shaker at 200rpm for 1 hour at 37°C in dark condition. Subsequently, the plate was centrifuged at 2500rpm for 5 minutes at RT followed by washing the pellet twice with 150 ⁇ l/well perm buffer. After every wash, centrifugation was carried out at 2500rpm for 5 minutes at RT. The cells were then re-suspended in 100 ⁇ l/well 1x PBS. The plate was then detected for cells in BD FACS Lyric and data was analysed using FlowJo software. The IC50 was calculated with respect to the virus control (cells treated with composition but not exposed to light).
  • Table 2 provides percentage inhibition of different concentration of the antiviral photosensitive composition (with light exposure) tested against different serotypes of the dengue virus.
  • composition (with light exposure) also demonstrated significant inhibitory activity against DENV infection, with IC50 values ranging from 0.03836 to 0.4419 ⁇ g/mL. This suggests that the composition is effective against all 4 serotypes of DENV.
  • percentage infectivity was plotted against the concentration of methylene blue in the composition using Graph pad Prism 8 to evaluate the IC50.
  • the X-axis plots the concentration of methylene blue in the composition, while the Y-axis shows the percentage of infection relative to the virus control (FIG. 7A and 7B).
  • the data was analyzed for nonlinear regression with Log inhibitor vs Normalized Response – Variable slope and the calculated IC50 from the same was presented.
  • FIG.7A A significant reduction in infection was observed when the composition was treated with light exposure (FIG.7A), indicating high antiviral potency; while a less pronounced reduction in infection was observed when the composition was not treated with light exposure, reflecting lower efficacy (FIG.7B).
  • Table 4 provides inhibitory activity (IC50) of the antiviral photosensitive composition (with and without light exposure, respectively) tested against different serotypes of the dengue virus.
  • Composition Inhibitory activity (IC50, ⁇ g/mL) DENV1 DENV2 DENV3 DENV4 Composition (With light exposure) 0.1005 0.03836 0.1297 0.4419 Composition (Without light exposure) 15.83 ⁇ 23.90 19.54 ⁇ 24.4
  • Cytotoxicity analysis by MTT assay [00103] On day 1, Vero cells were seeded at a concentration of 2.0 x 10 4 cells per well in a 96 well plate in 100 ⁇ l of complete media containing 10%FBS. The plate was incubated at 37°C with 5% CO2 for 24 hours.
  • the % viability and the % toxicity was plotted against concentration as shown in FIGs.8A and 8B. From the analysis the optimal concentration range of methylene blue was observed to be ranging from 0.78125 ⁇ g/mL to 1.5625 ⁇ g/mL. At this range, with light exposure, cell viability is maintained above 95 - 85%, and toxicity is minimal ( ⁇ 5 - 15%). Without light exposure, viability remains similarly high, offering a safe therapeutic window. Further, the concentration range (0.78125 ⁇ g/mL to 1.5625 ⁇ g/mL) minimizes toxicity while maintaining high cell viability, making it ideal for further testing or therapeutic applications.
  • FRNT Fluorescence Reduction Neutralization Test
  • the FRNT assay was performed to determine the ability of the antiviral photosensitive composition (with and without light exposure) to neutralize different serotypes of the dengue virus.
  • the assay included morphological observations of Vero cells treated with the antiviral photosensitive composition with light exposure (FIGs.9A and 9B) and without light exposure (FIGs.10A and 10B), under 20X magnification.
  • the images document changes in cellular health across different concentrations of the compound when infected with DENV-2.
  • Example 2 Redox potential calculation [00119] To calculate the redox potential generated by the photodynamic therapy (PDT) system involving methylene blue (MB), the energy delivered by the light sources has to be considered.
  • PDT photodynamic therapy
  • MB methylene blue
  • redox potential is an electrochemical property measured in volts, not directly derived from light intensity or exposure time.
  • Methylene blue has a high molar extinction coefficient around 610 nm, making it efficient at absorbing light.
  • Embodiments herein disclose an integrated solution for managing viral infections by combining methylene blue’s antiviral properties with a wearable platform (i.e., the device 100).
  • Embodiments herein disclose a non-invasive, broad-spectrum therapy that not only inhibits viral replication, but also modulates immune responses, reducing the risk of severe disease outcomes such as cytokine storms.
  • Embodiments herein disclose a scalable, patient-friendly solution for viral infections, with potential applications extending to other enveloped viruses and chronic conditions. By leveraging methylene blue’s safety, bioavailability, and efficacy, embodiments herein can serve as a next-generation therapeutic platform for infectious disease management.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Pathology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

An antiviral photosensitive composition and device for potentiating the same Embodiments herein disclose a device 100 for potentiating at least one phenothiazinium compound and method for operating the same. The device 100 is placed on a hand of the subject, such that the plurality of LEDs (205) are in proximity to the venous side of the hand of the subject. Embodiments herein further disclose an antiviral photosensitive composition including at least one phenothiazinium compound; at least one pharmaceutically acceptable carrier; and water. The antiviral photosensitive composition combined with the device 100, is effective against dengue virus and serotypes thereof.

Description

“An antiviral photosensitive composition and device for potentiating the same” CROSS REFERENCE TO RELATED APPLICATIONS This application is based on and derives the benefit of Indian Application IN 202341070238 filed on October 16, 2023, the contents of which are incorporated herein by reference in their entirety. TECHNICAL FIELD [001] The present invention relates to an antiviral photosensitive composition comprising a phenothiazinium compound, and more particularly to a device for potentiating the phenothiazinium compound for treatment of dengue virus (DENV) infections. BACKGROUND [002] Dengue is an acute febrile illness resulting from the infection caused by four serologically related positive-strand ribonucleic acid (RNA) viruses belonging to the Flavivirus genus, namely dengue viruses (DENV) 1, 2, 3, and 4. Annually, an estimated 390 million dengue infections occur globally, resulting in up to 36,000 fatalities. [003] It is primarily a mosquito-borne viral infection that can lead to a wide range of clinical manifestations and severity. DENV infection is generally characterized by high fever, retro orbital pain, myalgia, leukopenia, thrombocytopenia, and hemorrhagic manifestations. Majority of these cases recover within 4–7 days without requiring significant intervention. However, in some cases, dengue can progress to life threatening severe forms, such as Dengue Hemorrhagic Fever (DHF) or Dengue Shock Syndrome (DSS). Severe dengue is characterized by plasma leakage, severe bleeding, or organ dysfunction. Further, severe infections result in a significant clinical and economic burden due to the necessity for hospital admissions. [004] Given the urgent need to address the impact of advanced dengue infections, which can range from asymptomatic cases to more severe ones due to sequential infections by different dengue virus serotypes, the risk of developing severe dengue increases. Antibody-dependent enhancement (ADE) is a critical mechanism that can worsen the disease during secondary infections. In this process, a complex interaction between the virus and the host’s immune system intensifies the severity of the infection. This condition often escalates to a cytokine storm, pushing the patient into an irreversible state of distress, characterized by elevated cytokine levels and heightened immune activation. This, in turn, leads to increased vascular permeability and plasma leakage, which are key contributors to severe disease manifestations. [005] The existing treatments available in prior art are primarily symptomatic such as oral rehydration therapy and use of acetaminophen to alleviate fever or pain as per WHO recommendation 2009 policy. Currently, there is no approved antiviral medication for the treatment of dengue. Several clinical trials involving dengue patients have been conducted in recent years. Although none have demonstrated efficacy thus far, ongoing research and trials continue to explore potential treatments. Notably, many compounds tested in dengue clinical trials to date have been repurposed from molecules originally developed for other conditions. However, these efforts have largely failed to demonstrate effectiveness, highlighting the significant challenges in developing effective therapies for dengue. The complex relationship between viral load and disease severity further complicates the identification of effective therapeutic targets. [006] The key challenges today are the lack of specific antivirals, need for early diagnosis, and strengthening of healthcare capacity, while the focus of treatment remains on supportive care and managing complications. Novel therapeutic approaches targeting both the virus and host response are being actively looked upon. [007] In conclusion, the ongoing global burden of dengue underscores the urgent need for innovative solutions beyond symptomatic treatment. The lack of approved antiviral therapies leaves millions vulnerable to the severe complications of this disease. Advancing research in these areas is critical to developing comprehensive strategies that can effectively combat dengue, reduce its impact on affected populations, and ultimately save lives. OBJECTS [008] The principal object of embodiments herein is to disclose a device for potentiating at least one phenothiazinium compound in a user and method for operating the same, wherein the device is wearable on a wrist of the user and the process of potentiation is performed using a plurality of LEDs present on the underside of the device and in contact with venous side of the wrist of the user. [009] Another object of embodiments herein is to disclose an antiviral photosensitive composition. [0010] Another object of embodiments herein is to disclose an antiviral photosensitive composition effective against RNA viruses. [0011] Another object of embodiments herein is to disclose an antiviral photosensitive composition effective against dengue and serotypes thereof. [0012] Another object of embodiments herein is to disclose a method for treating infections caused by RNA viruses in a user. [0013] Another object of embodiments herein is to disclose a non-invasive treatment method for viral infections such as dengue, reducing the need for conventional antivirals. [0014] These and other objects of the embodiments herein will be better appreciated and understood when considered in conjunction with the following description and the accompanying drawings. It should be understood, however, that the following descriptions, while indicating preferred embodiments and numerous specific details thereof, are given by way of illustration and not of limitation. Many changes and modifications may be made within the scope of the embodiments herein without departing from the spirit thereof, and the embodiments herein include all such modifications. BRIEF DESCRIPTION OF DRAWINGS [0015] Embodiments herein are illustrated in the accompanying drawings, throughout which like reference letters indicate corresponding parts in the various figures. The embodiments herein will be better understood from the following description with reference to the following illustratory drawings. Embodiments herein are illustrated by way of examples in the accompanying drawings, and in which: [0016] FIG.1 depicts a device for potentiating methylene blue, according to embodiments as disclosed herein; [0017] FIG.2 depicts the components of the device, according to embodiments as disclosed herein; [0018] FIGs. 3A and 3B depict example layout of the LEDs on the device, according to embodiments as disclosed herein; [0019] FIG. 4 is a flowchart depicting the process of potentiating methylene blue using the device, according to embodiments as disclosed herein; [0020] FIG.5 is a flow diagram depicting the in vitro assay with VERO cells for testing the antiviral photosensitive composition, according to embodiments as disclosed herein; [0021] FIG.6 is a flow diagram depicting dilution preparation and plate layout for the in vitro assay, according to embodiments as disclosed herein; [0022] FIGs.7A and 7B are graphs depicting the IC50 values of the different concentrations of the antiviral photosensitive composition against different serotypes of the dengue virus (DENV 1-4), wherein FIG. 7A depicts % DENV infection when cells are treated with the antiviral photosensitive composition (with light exposure), and FIG.7B depicts % DENV infection when cells are treated with the antiviral photosensitive composition (without light exposure), according to embodiments as disclosed herein; [0023] FIGS. 8A and 8B are graphs depicting the percentage viability/toxicity of the Vero cells by MTT assay using the antiviral photosensitive composition, wherein FIG. 8A depicts the antiviral photosensitive composition (with light exposure), and FIG. 8B depicts the antiviral photosensitive composition (without light exposure), according to embodiments as disclosed herein; [0024] FIGs.9A and 9B are example cell morphology images from FRNT assay of Vero cells, wherein FIG.9A depicts example images of untreated cells (Cell and Virus control); and FIG.9B depicts example cells infected with DENV-2 and treated with different concentration of the antiviral photosensitive composition (with light exposure), according to embodiments as disclosed herein; and [0025] FIGs.10A and 10B are example cell morphology images from FRNT assay of Vero cells, wherein FIG. 10A depicts example images of untreated cells (Cell and Virus control); and FIG.10B depicts example cells infected with DENV-2 and treated with different concentration of the antiviral photosensitive composition (without light exposure), according to embodiments as disclosed herein. DETAILED DESCRIPTION [0026] The embodiments herein and the various features and advantageous details thereof are explained more fully with reference to the non-limiting embodiments that are illustrated in the accompanying drawings and detailed in the following description. Descriptions of well-known components and processing techniques are omitted so as not to unnecessarily obscure the embodiments herein. The examples used herein are intended merely to facilitate an understanding of ways in which the embodiments herein may be practiced and to further enable those of skill in the art to practice the embodiments herein. Accordingly, the examples should not be construed as limiting the scope of the embodiments herein. [0027] The words/phrases "exemplary", “example”, “illustration”, “in an instance”, “and the like”, “and so on”, “etc.”, “etcetera” are merely used herein to mean "serving as an example, instance, or illustration." Any embodiment or implementation of the present subject matter described herein using the words/phrases "exemplary", “example”, “illustration”, “in an instance”, “and the like”, “and so on”, “etc.”, “etcetera” is not necessarily to be construed as preferred or advantageous over other embodiments. [0028] The words/phrases "exemplary", “example”, “illustration”, “in an instance”, “and the like”, “and so on”, “etc.”, “etcetera”, “e.g.,”, “i.e.,” are merely used herein to mean "serving as an example, instance, or illustration." Any embodiment or implementation of the present subject matter described herein using the words/phrases "exemplary", “example”, “illustration”, “in an instance”, “and the like”, “and so on”, “etc.”, “etcetera”, “e.g.,”, “i.e.,” is not necessarily to be construed as preferred or advantageous over other embodiments. [0029] Embodiments herein may be described and illustrated in terms of blocks which carry out a described function or functions. These blocks, which may be referred to herein as managers, units, modules, hardware components or the like, are physically implemented by analog and/or digital circuits such as logic gates, integrated circuits, microprocessors, microcontrollers, memory circuits, passive electronic components, active electronic components, optical components, hardwired circuits and the like, and may optionally be driven by a firmware. The circuits may, for example, be embodied in one or more semiconductor chips, or on substrate supports such as printed circuit boards and the like. The circuits constituting a block may be implemented by dedicated hardware, or by a processor (e.g., one or more programmed microprocessors and associated circuitry), or by a combination of dedicated hardware to perform some functions of the block and a processor to perform other functions of the block. Each block of the embodiments may be physically separated into two or more interacting and discrete blocks without departing from the scope of the disclosure. Likewise, the blocks of the embodiments may be physically combined into more complex blocks without departing from the scope of the disclosure. [0030] It should be noted that elements in the drawings are illustrated for the purposes of this description and ease of understanding and may not have necessarily been drawn to scale. For example, the flowcharts/sequence diagrams illustrate the method in terms of the steps required for understanding of aspects of the embodiments as disclosed herein. Furthermore, in terms of the construction of the device, one or more components of the device may have been represented in the drawings by conventional symbols, and the drawings may show only those specific details that are pertinent to understanding the present embodiments so as not to obscure the drawings with details that will be readily apparent to those of ordinary skill in the art having the benefit of the description herein. Furthermore, in terms of the system, one or more components/modules which comprise the system may have been represented in the drawings by conventional symbols, and the drawings may show only those specific details that are pertinent to understanding the present embodiments so as not to obscure the drawings with details that will be readily apparent to those of ordinary skill in the art having the benefit of the description herein. [0031] The accompanying drawings are used to help easily understand various technical features and it should be understood that the embodiments presented herein are not limited by the accompanying drawings. As such, the present disclosure should be construed to extend to any modifications, equivalents, and substitutes in addition to those which are particularly set out in the accompanying drawings and the corresponding description. Usage of words such as first, second, third etc., to describe components/elements/steps is for the purposes of this description and should not be construed as sequential ordering/placement/occurrence unless specified otherwise. [0032] Embodiments herein disclose a device 100 for potentiating at least one phenothiazinium compound and method for operating the same. Referring now to the drawings, and more particularly to FIGs. 1 through 10B, where similar reference characters denote corresponding features consistently throughout the figures, there are shown embodiments. [0033] Embodiments herein use the terms ‘activation’, ‘activate’, ‘potentiate’, and so on interchangeably to refer to the process of enhancing the effect of a drug or a compound thereby allowing faster recovery. In an embodiment, the device 100 potentiates the antiviral properties of at least one phenothiazinium compound in an antiviral photosensitive composition. In an embodiment, the device 100 potentiates the antiviral properties of methylene blue in the antiviral photosensitive composition. [0034] The device 100, as depicted, can be in the form of a wearable device. In an embodiment herein, a person/subject can wear the device 100 on their wrist, wherein a bottom surface of the device 100 is placed in proximity to the underside of the person’s wrist (i.e., the venous side of the wrist) using a suitable means (such as, but not limited to, a strap). In the example depicted in FIG.1, the device 100 comprises a body 101 which can be at least one of temporarily or permanently attached to a strap 102. The strap 102 can be used to attach the device 100 to the wrist of a person securely, such that the underside of the device 100 is in contact with the underside of the wrist of the person (i.e., the venous side of the wrist of the person). Embodiments herein use the terms ‘user’, ‘subject’, ‘person’, and so on interchangeably to refer to the user on whose wrist to which the device is currently attached. [0035] In an embodiment herein, the device 100 does not comprise a strap, wherein the device 100 can be handheld against the underside of the person’s wrist (i.e., the venous side of the wrist) for a pre-defined period of time. [0036] The body 101 of the device 100, as depicted in FIG.2, comprises a controller 201, a timing module 202, a communication module 203, one or more user interfaces 204, a plurality of Light Emitting Diodes (LEDs) 205, at least one battery 206, and a battery charging module 207. [0037] The communication module 203 can enable one or more authorized users (such as a person administering the test/treatment, the person who is undergoing the test/treatment, a person who is supervising the test/treatment, and so on) to interact with the device 100. The communication module 203 can use one or more of a wired interface or a wireless interface to enable the one or more authorized users to interact with the device 100. Examples of the interface can be, but not limited to, Wi-Fi, a cellular network, Bluetooth, Bluetooth Low Energy (BLE), Near Field Communication (NFC), a Universal Serial Bus (USB) port, and so on. [0038] The one or more user interfaces 204 can enable the one or more authorized users to access and/or control the device 100. Examples of the one or more user interfaces 204 can be, but not limited to, one or more switches, toggles, a display, a touchscreen, a plurality of lights (such as LEDs (not the same as the LEDs 205)), a speaker, a microphone, a dial, and so on. [0039] The plurality of Light Emitting Diodes (LEDs) 205 can be placed on the bottom surface of the device 100, such as the LEDs are in proximity to the venous side of the wrist of the person, when the person is wearing the device 100. In an embodiment herein, the plurality of LEDs can be domed LEDs. In an embodiment herein, the plurality of LEDs can be non-domed LEDs. In an embodiment herein, the LEDs can have varying wavelengths. In an embodiment herein, the wavelength of the LEDs can range from 610 – 670 nm. In an embodiment herein, the number of LEDs 205 can comprise of two – twelve LEDs, arranged on the bottom surface of the device 100. In an example herein, the device 100 can comprise of six LEDs, wherein of the six LEDS, three LEDs can have a wavelength of 670nm (i.e., red light) with an intensity of 30000 – 50000Lux (+/ -5%), and the other three LEDs can have a wavelength of 610nm (i.e., amber light) with an intensity of 15000 – 25000Lux (+/ -5%). [0040] The redox potential of methylene blue (MB) is crucial when it interacts with light at 610nm and 670nm, as it determines how efficiently MB transitions between its oxidized and reduced forms. This characteristic plays a key role in several biological and therapeutic applications, particularly in photodynamic therapy (PDT), photoredox reactions, and antimicrobial treatments. Redox potential reflects the MB's ability to accept or donate electrons. When MB absorbs light, it can transition from an oxidized (MB⁺) state to a reduced state (leucomethylene blue, LMB) and back, depending on the environment and the available energy.610 nm light has slightly higher photon energy (~2.03 eV) as compared to 670 nm (~1.85 eV). This energy difference affects how efficiently MB undergoes photoexcitation and engages in redox cycling; the higher photon energy at 610nm can promote faster intersystem crossing (ISC) to triplet states, and the lower photon energy at 670nm can provide a more stable excitation with sufficient energy to trigger redox transitions, making it more suitable for using for a prolonged duration. [0041] In an embodiment herein, the LEDs 205 can be arranged in parallel with each other. In an embodiment herein, the LEDs 205 can be arranged in series with each other. In an embodiment herein, the LEDs 205 can be arranged in series and parallel with each other. In an example herein, the LEDs 205 can provide a light output of 1.2V – 2.4V. FIGs.3A and 3B depict example layouts of the LEDs on the device 100. [0042] The at least one battery 206 can provide power to the device 100, as required. In an example herein, the at least one battery 206 can be a Lithium-ion (Li-ion) battery. The at least one battery 206 can be charged by the battery charging module 207. The battery charging module 207 can use a suitable charging port for charging the battery 206. Examples of the charging port can be, but not limited to, a USB port, a micro-USB port, a USB-C port, a custom port, and so on. In an embodiment herein, the battery 206 can be charged using wireless charging. The battery charging module 207 can further a comprise a visual and/or an audio indicator (not shown), indicating the charging status (the device 100 is to be charged, the device 100 is charging, current battery level(s), the device 100 has completed charging, and so on). [0043] The term ‘controller 201,' as used in the present disclosure, may refer to, for example, hardware including logic circuits; a hardware/software combination such as a processor executing software; or a combination thereof. For example, the processing circuitry more specifically may include, but is not limited to, a central processing unit (CPU), an arithmetic logic unit (ALU), a digital signal processor, a microcomputer, a field programmable gate array (FPGA), a System-on- Chip (SoC), a programmable logic unit, a microprocessor, application-specific integrated circuit (ASIC), etc. For example, the controller 201 may include at least one of, a single processer, a plurality of processors, multiple homogeneous or heterogeneous cores, multiple Central Processing Units (CPUs) of different kinds, microcontrollers, special media, and other accelerators. [0044] Using the user interface 204, an authorized person can initiate the device operation for a pre-defined period of time. The pre-defined period of time can depend on the time required for a blood cell to complete one circulation through the subject (which depends on one or more physiological parameters of the user, such as, but not limited to, age of the user, physical condition of the user, health conditions of the user, blood flow rate of the user, and so on), wherein the pre- defined period of time can be equal to one – ten periods of time required for a blood cell to complete one circulation through the human body, and can be the time required for potentiating the at least one phenothiazinium compound. In an example herein, the pre-defined period of time can vary from 1 – 60 minutes. In an embodiment herein, the user can press a first switch to configure the device 100 to operate for a first period of time, the user can press a second switch to configure the device 100 to operate for a second period of time, and so on. In an embodiment herein, the user can use a touchscreen display to configure the device 100 to operate for a first period of time, the user can use a touchscreen display to configure the device 100 to operate for a second period of time, and so on. On receiving this intimation to initiate the device operation, the controller 201 can provide instructions to one or more LEDs to turn on/off for the configured pre- defined period of time. On receiving this intimation to initiate the device operation, in an embodiment herein, the controller 201 can provide instructions to all the LEDs 205 to light up for the pre-defined period of time. On receiving this intimation to initiate the device operation, in an embodiment herein, the controller 201 can provide instructions to a pre-defined set of LEDs 205 to light up for the pre-defined period of time standardized for adult & children respectively. The controller 201 can monitor the time of the device operation using the timing module 202. [0045] The LEDs 205 can turn ON and activate at least one phenothiazinium compound. In an embodiment herein, the at least one phenothiazinium compound can be methylene blue (MB). Light converts MB into a reduced form leucomethylene blue, a non-ionic compound that has the capability to modulate the immune system. The LEDs 205 can pass through skin at depths ranging from 3 to 4 mm. [0046] On the pre-configured period of time getting lapsed, the controller 201 can turn off the LEDs 205. In an embodiment herein, after turning off the LEDs 205, the controller 201 can provide an alert using a visual and/or an audio indicator (not shown) to the user that the device operation has been successfully completed. [0047] In an embodiment herein, the controller 201 can monitor the device operation for any errors/issues (such as, but not limited to, the device 100 not being in contact with the venous side of the wrist, the device 100 being removed/detaching from the wrist, low battery charge levels, the device 100 turning off (due to the battery getting fully discharged), and so on), and the controller 201 can provide an alert using a visual and/or an audio indicator (not shown) to the user about the error/issue. [0048] FIG.4 is a flowchart depicting the process of potentiating methylene blue. Using the user interface 204, in step 401, the authorized person (who can be at least one of a person authorized to perform the action, and the user to whose wrist the device is attached) initiates the device operation for the pre-defined period of time. On receiving this intimation to initiate the device operation, in step 402, the controller 201 provides instructions to the LEDs to turn ON for the configured pre-defined period of time. In step 403, the LEDs 205 turns ON. When the LEDs 205 are ON, in step 404, the least one phenothiazinium compound is potentiated immediately. On the pre-configured period of time lapsing (step 405), in step 406, the controller 201 turns off the LEDs 205. In an embodiment herein, after turning off the LEDs 205, the controller 201 provides an alert using a visual and/or an audio indicator (not shown) to the user that the device operation has been successfully completed. The various actions in method 400 may be performed in the order presented, in a different order or simultaneously. Further, in some embodiments, some actions listed in FIG.4 may be omitted. [0049] Embodiments herein further disclose a method for treating infections caused by RNA viruses in a subject. The method comprising administering an antiviral photosensitive composition including at least one phenothiazinium compound, at least one pharmaceutically acceptable carrier and water; and potentiating the least one phenothiazinium compound using the device 100. In an embodiment herein, the at least one phenothiazinium compound can be methylene blue. In an embodiment herein, the method includes administering the antiviral photosensitive composition, and potentiating methylene blue using the device 100. [0050] The term "user" or “subject”, as used interchangeably herein, refers to a member of an animal species of mammalian origin that may benefit from the administration of described antiviral photosensitive composition or method of the described invention. In an embodiment, the user is a human and animal. In an embodiment, the user is infected by RNA viruses. In an embodiment, the user is infected by viruses belonging to Flaviviridae family, including but not limited to, West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Feline Infectious Peritonitis, Zika virus, and serotypes thereof. In an embodiment, the virus is dengue virus and serotypes thereof. [0051] Embodiments herein disclose an antiviral photosensitive composition. The composition, according to embodiments herein includes at least one phenothiazinium compound; at least one pharmaceutically acceptable carrier selected from a group consisting of lipids, stabilizers, lubricants, and water. [0052] The term “photosensitive composition”, as used herein, refers to a mixture containing at least one compound capable of photoactivation, particularly when exposed to light, particularly visible light which potentiates the redox potential of the molecule to convert into ionic and nonionic form along with free oxygen liberation. It includes photosensitizing agents or photosensitizers, salts, or prodrug, or analogs, or derivatives thereof. Non limiting examples of photosensitizers include, prophyrins; dyes such as phenothiazinium, phenodiazinium, phenooxaziniums; chlorophyll; chlorins; bacteriochlorins; phenothiaziniums; prophycenes; and other natural occurring photosensitizing agents, or mixtures thereof. In an embodiment, the photosensitizer is a phenothiazinium compound. [0053] The composition according to embodiments herein includes at least one phenothiazinium compound. The terms “phenothiazinium” or “phenothiazines”, as used interchangeably herein, refers to a group of heterocyclic compounds having phototoxic efficiency against a broad range of microorganisms. Non-limiting examples of phenothiazinium compounds includes, Methylene blue (MB), 1-Octanol, 1,3- diphenylisobenzofuran (DPIBF), Rose Bengal (RB), new methylene blue (NMB), dimethyl methylene blue (DMMB), Azure A (AA), azure C (AC), azure B (AB), toluidine blue O (TBO), brilliant crystal blue (BCB), pyronin Y (PYY), neutral red (NR), derivatives and analogs thereof. In an embodiment, the phenothiazinium compound is methylene blue. [0054] Methylene Blue (MB) is a well-known tricyclic phenothiazinium compound with redox-active properties making it suitable for treating infectious diseases. It is approved by the FDA and EMA for the treatment of methemoglobinemia and malaria. As a redox-active agent, MB can oscillate between oxidized (MB⁺) and reduced (MBH) states. This redox cycling mechanism involves the reduction of MB to its leucomethylene blue form, which is then re-oxidized back to MB, creating a continuous cycle of redox reactions. The ionic form of MB is used to target the viral protease, while the nonionic form (leucomethylene blue) is involved in modulating cytokine storm & the redox cycling mechanism. [0055] Upon illumination, MB absorbs visible light energy and becomes activated, causing lipid envelope disintegration and RNA degradation in viruses, including those with double envelopes. These disrupt the viral membrane and cause destruction of the nucleic acids, particularly at guanosine residues. The resulting nucleic acid modification can prevent viral replication and induce the inactivation of viruses. [0056] In case of dengue, methylene blue disrupts the activity of NS1, a key viral protein essential for DENV replication and immune evasion. Further, MB interacts with proteins such as G3BP1, G3BP2, DDX6, Caprin1, and USP10, which are crucial for stress granule formation during DENV infection. This prevents viral replication by limiting stress granule assembly. Furthermore, MB regulates the unfolded protein response (UPR), activated under ER stress during infection, helping maintain ER homeostasis and reducing viral replication. [0057] MB also interferes with histone deacetylase (HDAC) activity, preventing chromatin condensation and restoring normal host transcription. It influences host gene expression by interacting with long non-coding RNAs (lncRNAs) and genes that promote viral replication through epigenetic regulation. Additionally, MB inhibits Positive Transcription Elongation Factor- b (P-TEFb), which helps reverse the overexpression of IL-8 associated with severe dengue hemorrhagic fever (DHF). By using an MB-based photodynamic therapy (PDT) platform, immune responses can be rebalanced, reducing the risk of cytokine storms. [0058] MB has poor penetration through cell membranes. Its decreased cellular uptake can be prevented by encapsulation in drug delivery systems such as liposomes. [0059] In an embodiment, the phenothiazinium compound, is present in an amount ranging from 0.001%w/w to 0.02%w/w including all the values in the range, for instance 0.002%w/w, 0.003%w/w, 0.004%w/w, 0.005%w/w, and so on. In an embodiment, methylene blue is present in an amount ranging from 0.001%w/w to 0.02%w/w. [0060] In an embodiment, the composition includes at least one pharmaceutically acceptable carrier. Non-limiting examples of pharmaceutically acceptable carriers includes solvents, diluents, lipids (fats and oils), binders, disintegrants, lubricants, preservatives, emulsifying agents, surfactants, stabilizers, humectants, buffers, flavoring and sweetening agents, coloring agents, thickening Agents (viscosity enhancers), coating agents, and so on. [0061] In an embodiment, the composition includes lipids. Non-limiting examples of lipids include phosphatidic acid, cholesterol, triglycerides such as caprylic/capric triglycerides, soybean oil, castor oil, olive oil; fatty acids such as linoleic acid, lauric acid, myristic acid, capric acid, arachidonic acid; fatty alcohols cetearyl alcohol, cetyl alcohol, oleyl alcohol, stearyl alcohol, lanolin alcohol; glycerides such as glyceryl monostearate, glyceryl distearate, glyceryl tricaprylate, glyceryl tristearate, glyceryl dibehenate; lecithin such as soy lecithin, egg lecithin; phospholipids such as lecithin, phosphatidylcholine (PC), non-hydrogenated phosphatidylcholine, phosphatidylglycerol (PG), phosphatidylserine (PS), Soya phosphatidylcholine (SPC), phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylinositol (PI), dioleoylphosphatidylcholine (DOPC), dimyristoylphosphatidylcholine (DMPC), dipentadecanoylphosphatidylcholine, dilauroylphosphatidylcholine (DLPC), soyaphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), distearoyl phosphatidylcholine (DSPC), diarachidonylphosphatidylcholine (DAPC), dihexanoyl phosphatidylcholine (DHPC), dioleoyl phosphatidylethanolamine (DOPE), dipalmitoylphosphatidylethanolamine (DPPE), distearoyl phosphatidylethanolamine (DSPE), dimyristoylphosphatidylserine(DMPS), distearoylphosphatidylserine (DSPS), dioleoylphosphatidylserine (DOPS), distearoylphosphatidylglycerol (DSPG), distearoylphosphatidic acid (DSPA), dipalmitoylphosphatidic acid (DPPA) or combinations thereof. In an embodiment, the lipid is non-hydrogenated phosphatidylcholine. In an embodiment, non-hydrogenated phosphatidylcholine acts as a binder in the present composition. [0062] In an embodiment, the lipid is present in an amount ranging from 3 %w/v to 4.5 %w/v including all the values in the range, for instance 3.1 %w/v, 3.2 %w/v, 3.3 %w/v, 3.4 %w/v, and so on. In an embodiment, non-hydrogenated phosphatidylcholine is present in an amount ranging from 3 %w/v to 4.5 %w/v. [0063] In an embodiment, the composition includes stabilizer. Non-limiting examples of stabilizers include antioxidants such as ascorbic acid, butylated hydroxytoluene, butylated hydroxyanisole, sodium metabisulfite, tocopherols; chelating agents such as ethylenediaminetetraacetic Acid (EDTA), disodium EDTA, citric acid; polyvinylpyrrolidone (PVP), hydroxypropyl Methylcellulose (HPMC), carboxymethylcellulose (CMC); amino acids such as arginine, histidine, glycine; sugars and polyols such as sucrose, sorbitol, mannitol, glycerin, and so on. In an embodiment, the stabilizer is glycerin. [0064] In an embodiment, the stabilizer is present in an amount ranging from 1 %w/v to 2.5 %w/v including all the values in the range, for instance 1.1 %w/v, 1.2 %w/v, 1.3 %w/v, 1.4 %w/v, and so on. In an embodiment, glycerin is present in an amount ranging from 1 %w/v to 2.5 %w/v. [0065] In an embodiment, the composition includes lubricants. Non-limiting examples of lubricants include magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, polyethylene glycol (PEG) or derivatives thereof, sodium benzoate, liquid paraffin, zinc stearate, calcium stearate, and so on. In an embodiment, the lubricant is polyethylene glycol (PEG) or derivatives thereof. Addition of PEG or PEG derivatives enhances the immunocompatibility of the active ingredient as PEGylated active ingredient exhibits prolonged in vivo circulation time resulting in their high bioavailability. Non-limiting examples of PEG and PEG derivatives include PEG 100, PEG 200, PEG 300, PEG 400, PEG 500, PEG 600, PEG 700, PEG 800, PEG 900, PEG 1000, PEG 1100, PEG 1200, PEG 1300, PEG 1400, PEG 1500, PEG 1600, PEG 1700, PEG 1800, PEG 1900, PEG 2000, PEG 2100, PEG 2200, PEG 2300, PEG 2400, PEG 2500, PEG 2600, PEG 2700, PEG 152800, PEG 2900, PEG 3000, PEG 3250, PEG 3350, PEG 3500, PEG 3750, PEG 4000, PEG 4250, PEG 4500, PEG 4750, PEG 5000, PEG 5500, PEG 6000, PEG 6500, PEG 7000, PEG 7500, PEG 8000, PEG 8500, PEG 9000, PEG 9500, PEG 10,000, PEG 11,000, PEG 12,000, PEG 13,000, PEG 14,000, PEG 15,000, PEG 16,000, PEG 17,000, PEG 18,000, PEG 19,000, PEG 20,000, mPEG (Methoxy Polyethylene Glycol), PEG-NHS (Polyethylene Glycol-N- hydroxysuccinimide), PEG-PLGA (Polyethylene Glycol-Polylactic-co-glycolic acid), PEG-DSPE (Polyethylene Glycol-Distearoyl Phosphoethanolamine), PEG-DMA (Polyethylene Glycol Dimethacrylate), PEG-PAA (Polyethylene Glycol-Polyacrylic Acid), PEG-Lactic Acid (PEG- PLA), PEG-Phospholipid (e.g., mPEG-Phosphatidylethanolamine), and so on. In an embodiment, the PEG is PEG 400. In an embodiment, the lubricant is PEG 400. [0066] In an embodiment, the lubricant is present in an amount ranging from 0.5 %w/v to 3 %w/v including all the values in the range, for instance 0.6 %w/v, 0.7 %w/v, 0.8 %w/v, 0.9 %w/v, and so on. In an embodiment, PEG 400 is present in an amount ranging from 0.5 %w/v to 3 %w/v. [0067] In an embodiment, the composition further includes water. The water is present in an amount sufficient to make 100%, i.e., water is in a quantity sufficient to make 100% of the composition. In an embodiment, the composition has a pH ranging from 5 to 6. [0068] Table 1 provides the antiviral photosensitive composition, according to embodiments herein. S. No. Ingredient Qty. 1 Phenothiazinium compound 0.001 to 0.02 %w/w 4 Lipid 3 to 4.5 %w/v 5 Stabilizer 1 to 2.5 %w/v 6 Lubricant 0.5 to 3 %w/v 7 Purified water, USP QS to 100% [0069] It is understood that various such modifications in the composition would be apparent to a person skilled in the art in light of the disclosures made herein and are included within the scope of the embodiments herein. [0070] Embodiment herein further disclose a method for preparing the antiviral photosensitive composition. In an embodiment, the method includes encapsulating the at least one phenothiazinium compound in liposomes. Various methods for preparing liposomes are generally known in the art which may be used in achieving the embodiments herein. These include, thin film hydration, ether/ethanol injection method, reverse phase evaporation method, detergent depletion method, heating method, microfluidic channel method, membrane extrusion method, homogenization and sonication method. In an embodiment, liposomes are prepared by homogenization and sonication method. [0071] The method for preparing the antiviral photosensitive composition, according to embodiments herein includes mixing at least one stabilizer in water to obtain a clear solution; dissolving at least one phenothiazinium compound in at least one solvent to obtain a phenothiazinium solution; mixing the clear solution, the phenothiazinium solution and adding at least one lipid; and sonicating and adding at least one lubricant to the resulting solution. [0072] In an embodiment, the method includes mixing at least one stabilizer in water to obtain a clear solution. Mixing may be achieved by any suitable method generally known in the field including, but not limited to, blending, sonicating, magnetic stirring, etc., by using equipment such as sonicator, magnetic stirrer, blender, mixer, etc. In an embodiment, mixing is performed for a period ranging from 5 to 15 minutes. In an embodiment, mixing is performed at 1300 to 1700 rpm. In an embodiment, the at least one stabilizer is mixed in water for a period ranging from 5 to 15 minutes at 1300 to 1700 rpm to obtain a clear solution. [0073] In an embodiment, the method includes dissolving at least one phenothiazinium compound in at least one solvent to obtain a phenothiazinium solution. Non-limiting examples of solvents include water, ethanol, chloroform, glacial acetic acid, glycerol, dimethyl formamide, and dimethyl sulfoxide. In an embodiment, the solvent is water. In an embodiment, methylene blue is dissolved in water. [0074] In an embodiment, the method further includes mixing the clear solution, the phenothiazinium solution and adding at least one lipid. In an embodiment, the lipid is non hydrogenated phosphatidylcholine. [0075] In an embodiment, the method further includes sonicating and adding at least one lubricant to the resulting solution. In an embodiment, sufficient quantity of water is added to make 100% of the composition before sonication. Sonication may be achieved by methods and conditions generally known in the art. In an embodiment, ultra-sonication is performed by passing the solution through a sonicator tunnel at a flow rate of 200 mL per minute, at 350 to 450 ultrasonic power. In an embodiment, sonication is performed for a time period ranging from 5 to 15 minutes. In an embodiment, the lubricant is polyethylene glycol. [0076] The composition may be filtered using methods generally known in the art. In an embodiment, the composition is filtered through 0.2 μm acetate cellulose membranes. [0077] The composition, according to embodiments herein, may further be formulated into any suitable dosage form. Non-limiting examples of dosage forms include, colloids, injectables, solution for oral or intravenous (IV)/ intramuscular (IM) administration, emulsions, suspensions, dispersion, capsules, tablets, lozenges, powder, lyophilized cakes, granules, gel, cream, lotions, and aerosols. In an embodiment, the dosage form of the composition is capsule. In one other embodiment, the dosage form of the composition is a tablet. Generally known methods of formulating/processing compositions may be used to formulate the desired dosage forms. In an embodiment, the disclosed composition is formulated in the form of capsule. In an embodiment, the disclosed composition is formulated in the form of tablets. [0078] The composition can be administered through various routes, including, but not limited to, oral, intravenous (IV), intramuscular (IM), subcutaneous (SC), topical, inhalation, and transdermal. In an embodiment, the dosage form is suited for oromucosal route of administration e.g.: buccal, sublingual, etc. In another embodiment, the composition is administered intraorally, and the components be prepared for uptake in a manner that makes the composition available in therapeutically effective amounts. As such, they may also be prepared as water soluble compositions, deliverable in encapsulated, or in a manner suitable for time release, delayed release or any manner typically used for delivery of pharmaceuticals, nutraceuticals or vitamins, etc. [0079] The dosage of the composition that may be used in the present invention will vary with the route of administration, the rate of excretion, the duration of the treatment, the identity of any other therapeutic compounds being administered, the age, size, and species of the subject, e.g., human patient, and like factors. In general, the dosage of a compound that may be used in the present invention will be an amount which is the lowest dose effective to produce the desired effect with minimal side effects or with very low side effect in comparison with therapeutic benefit. In an embodiment, the composition is administered in a dosage ranging from 1 to 15 mg per day which can be administered in single or multiple doses. [0080] In an embodiment, the device 100 combined with the antiviral photosensitive composition, is effective against DENV and other RNA viruses. Non-limiting examples of RNA viruses includes, Coronavirus family (Coronaviridae), Flavivirus family (Flaviviridae) such as West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Zika virus, and serotypes thereof; Orthomyxovirus family (Orthomyxoviridae) such as Influenza A, B, and C viruses; Retrovirus family (Retroviridae); Filovirus family (Filoviridae); Paramyxovirus family (Paramyxoviridae); Togavirus family (Togaviridae); and Picornavirus family (Picornaviridae). [0081] In an embodiment, the device 100 combined with the antiviral photosensitive composition are effective against dengue virus and serotypes thereof. In an embodiment, the dengue virus is DENV-1. In one other embodiment, the dengue virus is DENV-2. In yet one other embodiment, the dengue virus is DENV-3. In one other embodiment, the dengue virus is DENV- 4. [0082] The inventors of the present invention have achieved a methylene blue-based composition-device combination therapy where the redox potential of MB is exploited using photodynamic therapy (PDT). PDT further enhances MB’s effect by potentiating redox potential converting into ionic and nonionic form, maximizing viral inactivation. The device 100 potentiates MB at 610-670 nm, generating redox cycle amplification of MB at the application site. MB cycles between oxidized and reduced states to modulate cellular oxidative stress, disrupting viral replication. Thus, the methylene blue-based composition-device combination therapy provides an enhanced antiviral activity through combined form (ionic and non-ionic form) of molecule. The therapy also targets the host body by reducing oxidative stress and inflammation, which are common complications of dengue virus infections. The device 100 further provides precise wavelength activation ensuring efficient MB activation with minimal phototoxicity. [0083] The inventors of the present invention further achieved an effective composition against dengue virus with even a low concentration (i.e.,1 to 2 mg) of methylene blue, thereby minimizing side effects of methylene blue. Further, the disclosed composition is capable of bypassing first pass metabolism. The composition can overcome potential loss or reduction in active ingredient i.e., methylene blue caused due to first pass effect or pre-systemic metabolism, thereby improving its bioavailability and stability. Further the present invention contributes to antimicrobial resistance (AMR) mitigation by reducing dependence on traditional antivirals. [0084] Embodiments are further described herein by reference to the following examples by way of illustration only and should not be construed to limit the scope of the claims provided herewith. Example 1: Preparation of the antiviral photosensitive composition [0085] Initially 12,500 mg glycerin (USP or BP Grade) was dissolved in 500ml purified water. The mixing was carried out at 1500 rpm for 10 minutes under room temperature to obtain a clear solution. Subsequently, a solution of methylene blue was prepared by dissolving 10000mg methylene blue in 100ml ml of purified water. Both the glycerin solution and the methylene blue solution were mixed along with addition of 15000mg of non-hydrogenated phosphatidylcholine solution. Purified water was added to the resulting solution to bring the total volume to 900 mL. [0086] Later, the solution was sonicated using ultra sonicate probe by passing the solution through a sonicator tunnel at 200ml /min with 400UP (ultrasonic power) for 10 minutes. Subsequently, 1g/100ml of polyethylene glycol was added to the solution. The final volume was adjusted, and the composition was finally filtered through 0.2 μm acetate cellulose membranes. [0087] The inventors of the present invention conducted a series of experiments to determine the optimal concentration of the phenothiazinium compound (methylene blue) in the antiviral photosensitive composition (along with light exposure) for maximum efficacy against different serotypes of the dengue virus. Experiment 1: Screening of the composition against DENV1-4 [0088] To test the antiviral photosensitive composition, Vero cells were used. The Vero cell line, derived from the kidney epithelial cells of the African Green Monkey, is highly susceptible to a wide range of viral infections, including many human and animal viruses. This characteristic makes them a versatile tool for studying and screening antiviral compounds against various types of viruses, including Dengue Virus (DENV). In addition to this, Vero cells do not produce interferon which help researchers to study effectiveness of antiviral compound. [0089] On day 1, 2.5 x 10⁵ Vero cells were seeded per well in a 96-well plate with 100 µl of complete DMEM medium containing 5% ΔFBS (Fetal bovine serum). The cells were then incubated in a humidified incubator at 37°C with 5% CO₂ for 24 hours to allow them to adhere and attain proper morphology. [0090] On Day 2, the cells were infected with each DENV strain separately at an MOI (Multiplicity of Infection) of 0.1 (5000 FIU (Fluorescent Infectious Units) of each virus). The infection was carried out in 40 µl of infection media per well, consisting of 1x DMEM with 2% ΔFBS, and the cells were incubated for 2 hours in a humidified incubator at 37°C with 5% CO₂. After 2 hrs, the virus containing media was aspirated from the wells, followed by washing the well with 100µl incomplete (1x DMEM) media. [0091] After washing the overlayed media, the cells were treated with different concentration of the antiviral photosensitive composition along with light exposure. FIG. 5 is a flow diagram depicting the in vitro assay using VERO cells to test the antiviral photosensitive composition. [0092] The composition was further diluted by dissolving in media (DMEM + 2% FBS) to obtain a concentration of 100 ug/mL. For determining the effective concentration of methylene blue, different dilutions (dilution ranging from 100 µg/mL to 0.1953 µg/mL) in media were further prepared (FIG.6). Treatment of cells with the composition [0093] Culture media was aspirated and 100µl/well of the disclosed composition was added from the dilution plate in duplicates. After treatment, the cells were exposed to 20 minutes of light. Post light exposure, the plate was incubated further for another 48 hours in humidified incubator at 37°C with 5% CO2. Another set of DENV1-4 infected plates containing the disclosed composition was prepared, where the cells were not exposed to light. FACS Staining [0094] On day 4, the cells were observed, and the media was aspirated and washed with 150 µl/ well of 1X PBS (phosphate buffered saline). Subsequently, the cells were trypsinised by adding 25µl/well Trypsin-EDTA solution. Further, to inactivate the trypsin, 150µl/well 10% FBS (in 1x PBS) was added followed by mixing and gently transferring the cells to a 96 well U-bottom plate. [0095] The plate was then centrifuged at 1500 rpm for 5 minutes at room temperature (RT). After centrifugation, the supernatant was aspirated, and the pellet was washed with 150 µl/well of 1X PBS. The plate was then centrifuged again at 1500 rpm for 5 minutes at RT. Following this, the supernatant was aspirated, and the cells were fixed by resuspending the pellet in 50 µl/well of 4% paraformaldehyde (PFA). The plate was incubated for 10 minutes at RT to allow fixation. Followed by centrifugation at 2500 rpm for 5 minutes at RT. The pellet was further washed twice with 150 µl/well of 1X Permeabilization Buffer (perm buffer). After every wash, centrifugation was carried out at 2500rpm for 5 minutes at RT. The cells were then treated with 40 µl/ well Blocking solution (1% Normal Mouse Serum in 1x Perm Buffer) followed by re-suspending the cells and incubating the plate at RT for 30 mins.20 µl/ well Alexa Fluor 488 labelled anti-DENV monoclonal antibody was then added along with blocking solution. [0096] The cells were then mixed and incubated in a plate shaker at 200rpm for 1 hour at 37°C in dark condition. Subsequently, the plate was centrifuged at 2500rpm for 5 minutes at RT followed by washing the pellet twice with 150µl/well perm buffer. After every wash, centrifugation was carried out at 2500rpm for 5 minutes at RT. The cells were then re-suspended in 100µl/well 1x PBS. The plate was then detected for cells in BD FACS Lyric and data was analysed using FlowJo software. The IC50 was calculated with respect to the virus control (cells treated with composition but not exposed to light). Controls were run parallelly and were treated similarly: Mock control [cell control: neither infected nor treated); Virus control: Cells were infected with each one of DENV serotype, separately, in the absence of compound]. The concentration of the compound (methylene blue) that caused a 50% reduction in the number of DENV-infected cells, relative to the virus control representing 100% infection, was designated as the 50% inhibitory concentration (IC50) of the tested compounds. The IC50 value was calculated through GraphPad prim (version 8) and graph was plotted against each DENV serotypes. Percentage Inhibition against DENV 1-4 [0097] A significant cell mortality at both the starting concentrations i.e,100 µg/mL and 50 µg/mL was observed. Because of this, cells treated with these two initial concentrations were found to be not suitable for flow cytometry analysis. Consequently, the data from these concentrations was not included in the subsequent results. [0098] Table 2 provides percentage inhibition of different concentration of the antiviral photosensitive composition (with light exposure) tested against different serotypes of the dengue virus. Comp % Inhibition with respect to Virus control conc.(µg) DENV 1 DENV 2 DENV 3 DENV 4 25 94.33 96.03 94.17 90.71 97 96.75 92.31 84.63 12.5 97.59 96.05 95.51 97.7 97.58 98.14 95.2 88.48 6.25 89.86 95.18 93.02 97.09 95.23 96.78 80.15 63.57 3.125 68.88 89.35 88.9 92.94 93.15 93.42 77.06 55.38 1.5625 61.23 72.91 69.77 80.89 84.71 83.22 58.11 45.64 0.78125 51.08 62.13 63.43 75.95 78.56 69.57 63.96 53.84 0.390625 65.14 79.25 72 88.43 84.08 77.63 77.67 53.84 0.1953125 52.15 73.1 63.76 71.77 48.82 57.1 59.39 29.7 [0099] Table 3 provides percentage inhibition of different concentration of the antiviral photosensitive composition (without any light exposure) tested against different serotypes of the dengue virus. Comp % Inhibition with respect to Virus control conc. (µg) DENV 1 DENV 2 DENV 3 DENV 4 25 84.9 80.77 83.48 75.51 80.63 73.53 67.22 67.47 12.5 45.9 16.39 -20.07 -49.86 15.93 10 -45.88 -66.55 6.25 36.72 -36.01 -94.03 -117.7 -49.53 -58.03 -138.8 -70.34 3.125 12.18 -28.24 -63.36 -106.8 -36.83 -38.27 -101.3 -92.53 1.5625 -24.92 -17.68 -49.88 -73.61 -9.71 -35.52 -33.97 -53.57 0.78125 -6.56 -15.74 -15.77 -56.65 -11.95 -15.01 -17.88 -53.03 0.390625 -15.36 -10.13 -7.74 -26.12 -16.18 -14.76 0 -28.14 0.1953125 8.43 -9.7 -12.61 -17.98 -15.43 -7.5 6.55 -15.69 [00100] From the Table 2 and 3, it can be observed that the antiviral photosensitive composition tested against Dengue Virus (DENV) serotypes 1- 4, showed significant inhibitory activity against DENV infection. The composition (with light exposure) also demonstrated significant inhibitory activity against DENV infection, with IC50 values ranging from 0.03836 to 0.4419 μg/mL. This suggests that the composition is effective against all 4 serotypes of DENV. [00101] Further, percentage infectivity was plotted against the concentration of methylene blue in the composition using Graph pad Prism 8 to evaluate the IC50. The X-axis plots the concentration of methylene blue in the composition, while the Y-axis shows the percentage of infection relative to the virus control (FIG. 7A and 7B). The data was analyzed for nonlinear regression with Log inhibitor vs Normalized Response – Variable slope and the calculated IC50 from the same was presented. A significant reduction in infection was observed when the composition was treated with light exposure (FIG.7A), indicating high antiviral potency; while a less pronounced reduction in infection was observed when the composition was not treated with light exposure, reflecting lower efficacy (FIG.7B). [00102] Table 4 provides inhibitory activity (IC50) of the antiviral photosensitive composition (with and without light exposure, respectively) tested against different serotypes of the dengue virus. Composition Inhibitory activity (IC50, µg/mL) DENV1 DENV2 DENV3 DENV4 Composition (With light exposure) 0.1005 0.03836 0.1297 0.4419 Composition (Without light exposure) 15.83 ~23.90 19.54 ~24.4 Experiment 2: Cytotoxicity analysis by MTT assay [00103] On day 1, Vero cells were seeded at a concentration of 2.0 x 104 cells per well in a 96 well plate in 100µl of complete media containing 10%FBS. The plate was incubated at 37°C with 5% CO2 for 24 hours. On day 2, different dilution of the antiviral photosensitive composition were prepared in a media containing DMEM and 2% FBS to obtain a concentration of 25 ug/mL. (Stock conc: 5 mg/ml). Different dilutions of the composition were further prepared in media ranging from 25 µg to 0.195 µg. Treatment of cells with the composition [00104] Culture media was aspirated and 100µl/well of the disclosed composition was added from the dilution plate in duplicates. After treatment, the cells were exposed to 20 minutes of light. Post light exposure, the plate was incubated further for another 48 hours in humidified incubator at 37°C with 5% CO2. Another set containing the disclosed composition was prepared, where the cells were not exposed to light. [00105] After 48 hours, 10µl of 5mM MTT solution (made in 1X PBS) was added in each well in dark conditions. The plates were further incubated for 4 hours at 37° C till the formation of formazan crystals. 100µl of isopropanol was added in each well followed by incubation at 37 °C in shaking condition for 15-30 mins till the crystals were observed to dissolve completely. The plate was observed at 575nm in UV-Vis Spectrophotometer, with MTT and Isopropanol/DMSO mixture considered as blank [00106] The reading is proportional to the live cells. The viability and toxicity were calculated in percentage by the following formula: ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ − ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ % = ( ) × 100 ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ℎ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ − ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ Toxicity % = 100 – Viability % [00107] The viability% and toxicity% was plotted against concentration and the intersection point of both formed at 50%. The concentration point at this intersection was depicted as CC50. The study evaluates the effect of an antiviral photosensitive composition on Vero cells under two conditions: with and without light exposure. [00108] Table 5 provides the percentage viability of Vero cells treated with the antiviral photosensitive composition (with 20 minutes light exposure). Conc. % Viability % Toxicity 25 68.27306 68.4358 31.72694 31.5642 12.5 64.86438 62.10669 35.13562 37.89331 6.25 67.54069 65.70524 32.45931 34.29476 3.125 62.96564 79.54792 37.03436 20.45208 1.5625 85.92224 84.50271 14.07776 15.49729 0.78125 95.17179 85.50633 4.82821 14.49367 0.390625 95 92.72152 5 7.278481 0.195313 88.02893 97.48644 11.97107 2.513562 [00109] Table 6 provides the percentage viability of Vero cells treated with the antiviral photosensitive composition (without light exposure). Conc. % Viability % Toxicity 25 84.19038 81.33464 15.80962 18.66536 12.5 91.24632 82.87537 8.75368 17.12463 6.25 94.78901 85.44652 5.210991 14.55348 3.125 97.25221 98.72424 2.747792 1.275761 1.5625 89.32287 88.82237 10.67713 11.17763 0.78125 96.32974 94.9264 3.670265 5.073602 0.390625 94.37684 94.94603 5.62316 5.053974 0.195313 97.36016 98.16487 2.639843 1.835132 [00110] From Table 5 it can be observed that higher concentration of the composition (i.e., 25, 12.5 µg/mL; with light exposure) resulted in lower cell viability (~62-68%) and higher toxicity (~31-37%). As concentration decreases, viability increases steadily (up to 95% at 0.78125 µg/mL), and toxicity declines. The lowest concentrations (0.390625 µg/mL and 0.195313 µg/mL) showed high viability (~88-95%) and minimal toxicity. [00111] From Table 6 it can be observed that higher concentrations of the composition (without light exposure) also showed better outcomes than with light exposure. For instance, at 25 µg/mL, cell viability was found to be ~84% much higher compared to the light-exposed group. At lower concentrations (3.125-0.195313 µg/mL), viability remained above 94%, with toxicity decreasing below 5%. Cells treated without light exposure were observed to be generally more viable across all concentrations compared to those exposed to light. [00112] Overall, it was observed that light exposure reduces cell viability, especially at higher concentrations, suggesting potential photo-toxicity. Both with and without light exposure, lower concentrations yield better viability, implying an existence of optimal dose range. These results highlighted the importance of balancing concentration and light exposure for effective use of the antiviral composition. [00113] Further, percentage viability/toxicity was plotted against the concentration of methylene blue in the composition using Graph pad Prism 8 to evaluate the CC50 values. The X- axis plots the concentration of methylene blue in the composition, while the Y-axis shows the percentage of viability and toxicity. The % viability and the % toxicity was plotted against concentration as shown in FIGs.8A and 8B. From the analysis the optimal concentration range of methylene blue was observed to be ranging from 0.78125 µg/mL to 1.5625 µg/mL. At this range, with light exposure, cell viability is maintained above 95 - 85%, and toxicity is minimal (~5 - 15%). Without light exposure, viability remains similarly high, offering a safe therapeutic window. Further, the concentration range (0.78125 µg/mL to 1.5625 µg/mL) minimizes toxicity while maintaining high cell viability, making it ideal for further testing or therapeutic applications. Experiment 3: FRNT (Focus Reduction Neutralization Test) assay [00114] The FRNT assay (Focus Reduction Neutralization Test) was performed to determine the ability of the antiviral photosensitive composition (with and without light exposure) to neutralize different serotypes of the dengue virus. The assay included morphological observations of Vero cells treated with the antiviral photosensitive composition with light exposure (FIGs.9A and 9B) and without light exposure (FIGs.10A and 10B), under 20X magnification. The images document changes in cellular health across different concentrations of the compound when infected with DENV-2. [00115] At high concentrations of the composition with light exposure (50-100 µg/mL), notable cell stress and damage were evident, with fewer healthy cells present. At moderate concentrations with light exposure (6.25-25 µg/mL), the cells appeared healthier, showing some viral inhibition and indicating partial protection. At low concentrations with light exposure (≤3.125 µg/mL), cell morphology was mostly preserved, with minimal signs of cytotoxicity or infection (FIG.9B). [00116] For cells treated with the antiviral photosensitive composition without light exposure, high concentrations (50-100 µg/mL), resulted in similar stress but with more pronounced viral damage compared to light-exposed samples. At moderate to low concentrations, the cells appeared more infected and damaged, indicating reduced efficacy of the composition without light activation. [00117] In the case of controls (FIGs. 9A and 10A), the virus control shows infected cells with widespread damage and altered morphology, while the cell control, consisting of healthy untreated cells, retained normal morphology and structure. [00118] Overall, it was observed that the antiviral photosensitive composition, when used in conjunction with light exposure, was more effective in reducing viral damage and maintaining healthier cell morphology, particularly at moderate concentrations. Example 2: Redox potential calculation [00119] To calculate the redox potential generated by the photodynamic therapy (PDT) system involving methylene blue (MB), the energy delivered by the light sources has to be considered. However, redox potential is an electrochemical property measured in volts, not directly derived from light intensity or exposure time. The key parameters relevant here are: - Energy of light absorbed by MB (depends on wavelength and intensity) - Excitation and subsequent oxygen species generation efficiency for methylene blue; and - Absorption and extinction coefficient of methylene blue at 610 nm and 670 nm. Energy calculation: [00120] Using the formula for energy: ^^ = ^^. ^^ Where ^^ = ^^ ^^ ^^ ^^ ^^ ^^ ( ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^) ^^ = ^^ ^^ ^^ ^^ ^^ ( ^^ ^^ ^^ ^^ ^^ ^^ ^^, ^^ℎ ^^ ^^ℎ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ × ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^) t = Time (in seconds) [00121] Since 1 Lux = 1 Lumen per square meter (assuming isotropic uniform lighting), the power contribution from the LEDs can be integrated over the total exposure time. [00122] For the wavelength-specific energy per photon, ℎ ^^ ^^ ^^ℎ ^^ ^^ ^^ ^^ = ^^ Where
Figure imgf000030_0001
ℎ ^^ ^^ ^^ℎ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ (6.626 × 10−34 ^^. ^^) ^^ ^^ ^^ ^^ℎ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ℎ ^^ (3 × 108 ^^/ ^^) ^^ ^^ ^^ ^^ℎ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^ℎ ( ^^ ^^ ^^ ^^ ^^ ^^ ^^ ^^) For 610 nm (610 × 10⁻⁹ m): (6.626 × 10−34) × (3 × 108) ^^ ^^ℎ ^^ ^^ ^^ ^^−610 = = 3.25 × 10−19 ^^ 610 × 10−9 For 670 nm (670 × 10⁻⁹ m): (6.626 × 10−34) × (3 × 108) ^^ ^^ℎ ^^ ^^ ^^ ^^−670 = = 2.97 × 10−19 ^^ 670 × 10−9 Power Contribution from LEDs: [00123] In the current example, consider that there are 3 LEDs at 610 nm with 19000 Lux each, and 3 LEDs at 670 nm with 35000 Lux each. The total Lux contribution from each set of LEDs is: ^^610 = 3 × 19000 = 57000 ^^ ^^ ^^ ^^670 = 3 × 35000 = 105000 ^^ ^^ ^^ [00124] Since Lux measures light intensity per unit area, the total power across the treatment zone (say 1 m²) can be approximated to 57 W/m² for 610 nm and 105 W/m² for 670 nm. [00125] Consider that the pre-defined time period is 50 minutes, the energy delivered for each wavelength in 50 minutes (i.e., 3000 seconds) is ^^610 = 57 ^^⁄ ^^2 × 3000 ^^ ^^ ^^ ^^ ^^ ^^ ^^ = 171000 ^^⁄ ^^2 ^^670 = 105 ^^ ^^2 × 3000 ^^ ^^ ^^ ^^ ^^ ^^ ^^ = 315000 ^^ ^^2 [00126] Methylene blue has a high molar extinction coefficient around 610 nm, making it efficient at absorbing light. At 5 mg (assuming molar mass ~ 319.85 g/mol), the concentration is approximately: 5 ^^ ^^ = 1.56 × 10−5 ^^ ^^ ^^ 319.85 ^^ ^^ ^^ ^^ [00127] If all the absorbed photons are used to potentiate the methylene blue, the redox cycle amplification potential generated will be influenced by the photon absorption, MB concentration, light intensity & duration time. [00128] The energy delivered by the 610 nm and 670 nm light sources totals around 486,000 J/m² over 50 minutes. This energy, absorbed by methylene blue, initiates redox reactions amplification and generates oxygen species, influencing biological targets during photodynamic therapy. [00129] Embodiments herein disclose an integrated solution for managing viral infections by combining methylene blue’s antiviral properties with a wearable platform (i.e., the device 100). Embodiments herein disclose a non-invasive, broad-spectrum therapy that not only inhibits viral replication, but also modulates immune responses, reducing the risk of severe disease outcomes such as cytokine storms. [00130] Embodiments herein disclose a scalable, patient-friendly solution for viral infections, with potential applications extending to other enveloped viruses and chronic conditions. By leveraging methylene blue’s safety, bioavailability, and efficacy, embodiments herein can serve as a next-generation therapeutic platform for infectious disease management. [00131] The foregoing description of the specific embodiments will so fully reveal the general nature of the embodiments herein that others can, by applying current knowledge, readily modify and/or adapt for various applications such specific embodiments without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. Therefore, while the embodiments herein have been described in terms of preferred embodiments, those skilled in the art will recognize that the embodiments herein can be practiced with modification within the spirit and scope of the embodiments as described herein.

Claims

CLAIMS We claim: 1. A device (100) comprising: a controller (101); and a plurality of Light Emitting Diodes (LEDs) (205) on a bottom surface of the device (100), wherein the controller (101) is configured to: potentiate at least one phenothiazinium compound in a subject by turning ON at least one of the plurality of LEDs (205) for a pre-defined period of time, wherein the device is placed on a hand of the subject, such that the plurality of LEDs (205) are in proximity to the venous side of the hand of the subject.
2. The device, as claimed in claim 1, wherein at least one LED of the plurality of LEDs (205) has a wavelength of 670nm with an intensity of 30000 – 50000Lux (+/ -5%), and at least one LED of the plurality of LEDs (205) has a wavelength of 610nm with an intensity of 15000 – 25000Lux (+/ -5%).
3. The device, as claimed in claim 1, wherein at least one LED of the plurality of LEDs (205) has a light output of 1.2V-2.4V.
4. The device, as claimed in claim 1, wherein the pre-defined period of time can depend on a time required for a blood cell to complete one circulation through the subject.
5. The device, as claimed in claim 1, wherein the device (100) further comprises a strap (102), a timing module (202), a communication module (203), one or more user interfaces (204), at least one battery (206), and a battery charging module (207).
6. An antiviral photosensitive composition, comprising at least one phenothiazinium compound; at least one pharmaceutically acceptable carrier selected from a group consisting of lipids, stabilizers, and lubricants; and water, wherein the least one phenothiazinium compound is potentiated by the device 100 as claimed in claim 1.
7. The composition as claimed in claim 6, wherein the at least one phenothiazinium compound is selected from a group consisting of Methylene blue (MB), 1-Octanol, 1,3- diphenylisobenzofuran (DPIBF), Rose Bengal (RB), new methylene blue (NMB), dimethyl methylene blue (DMMB), Azure A (AA), azure C (AC), azure B (AB), toluidine blue O (TBO), brilliant crystal blue (BCB), pyronin Y (PYY), neutral red (NR), derivatives and analogs thereof.
8. The composition as claimed in claim 7, wherein the at least one phenothiazinium compound is methylene blue.
9. The composition as claimed in claim 6, wherein the at least one phenothiazinium compound is present in an amount ranging from 0.001%w/w to 0.02%w/w.
10. The composition as claimed in claim 6, wherein the at least one pharmaceutically acceptable carrier is a lipid selected from a group consisting of phosphatidic acid, cholesterol, triglycerides, linoleic acid, lauric acid, myristic acid, capric acid, arachidonic acid, cetearyl alcohol, cetyl alcohol, oleyl alcohol, stearyl alcohol, lanolin alcohol, glyceryl monostearate, glyceryl distearate, glyceryl tricaprylate, glyceryl tristearate, glyceryl dibehenate, soy lecithin, egg lecithin, lecithin, phosphatidylcholine (PC), non-hydrogenated phosphatidylcholine, phosphatidylglycerol (PG), phosphatidylserine (PS), soya phosphatidylcholine (SPC), phosphatidylethanolamine (PE), phosphatidic acid (PA), phosphatidylinositol (PI), dioleoylphosphatidylcholine (DOPC), dimyristoylphosphatidylcholine (DMPC), dipentadecanoylphosphatidylcholine, dilauroylphosphatidylcholine (DLPC), soyaphosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC), distearoyl phosphatidylcholine (DSPC), diarachidonylphosphatidylcholine (DAPC), dihexanoyl phosphatidylcholine (DHPC), dioleoyl phosphatidylethanolamine (DOPE), dipalmitoylphosphatidylethanolamine (DPPE), distearoyl phosphatidylethanolamine (DSPE), dimyristoylphosphatidylserine(DMPS), distearoylphosphatidylserine (DSPS), dioleoylphosphatidylserine (DOPS), distearoylphosphatidylglycerol (DSPG), distearoylphosphatidic acid (DSPA), dipalmitoylphosphatidic acid (DPPA), and derivatives thereof.
11. The composition as claimed in claim 10, wherein the lipid is non-hydrogenated phosphatidylcholine.
12. The composition as claimed in claim 10, wherein the lipid is present in an amount ranging from 3 %w/v to 4.5 %w/v.
13. The composition as claimed in claim 6, wherein the at least one pharmaceutically acceptable carrier is a stabilizer selected from a group consisting of ascorbic acid, butylated hydroxytoluene, butylated hydroxyanisole, sodium metabisulfite, tocopherols, ethylenediaminetetraacetic Acid (EDTA), disodium EDTA, citric acid, polyvinylpyrrolidone (PVP), hydroxypropyl methylcellulose (HPMC), carboxymethylcellulose (CMC), arginine, histidine, glycine, sucrose, sorbitol, mannitol, glycerin, and derivatives thereof.
14. The composition as claimed in claim 13, wherein the stabilizer is glycerin.
15. The composition as claimed in claim 13, wherein the stabilizer is present in an amount ranging from 1 %w/v to 2.5 %w/v.
16. The composition as claimed in claim 6, wherein the at least one pharmaceutically acceptable carrier is a lubricant selected from a group consisting of magnesium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, polyethylene glycol (PEG), sodium benzoate, liquid paraffin, zinc stearate, calcium stearate, and derivatives thereof.
17. The composition as claimed in claim 16, wherein the lubricant is polyethylene glycol (PEG) or derivative thereof.
18. The composition as claimed in claim 15, wherein the lubricant is present in an amount ranging from 0.5 %w/v to 3 %w/v.
19. A method for treating infections caused by RNA viruses in a subject comprising: administering an antiviral photosensitive composition comprising at least one phenothiazinium compound, at least one pharmaceutically acceptable carrier selected from a group consisting of lipids, stabilizers, lubricants, and water; and potentiating at least one phenothiazinium compound using the device 100 as claimed as claim 1.
20. The method as claimed in claim 19, wherein the RNA viruses are selected from a group consisting of Coronavirus family (Coronaviridae), Flavivirus family (Flaviviridae) such as West Nile virus, dengue virus, tick-borne encephalitis virus, yellow fever virus, Zika virus, and serotypes thereof; Orthomyxovirus family (Orthomyxoviridae) such as Influenza A, B, and C viruses; Retrovirus family (Retroviridae); Filovirus family (Filoviridae); Paramyxovirus family (Paramyxoviridae); Togavirus family (Togaviridae); and Picornavirus family (Picornaviridae).
PCT/IN2024/052076 2023-10-16 2024-10-16 An antiviral photosensitive composition and device for potentiating the same Pending WO2025083710A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN202341070238 2023-10-16
IN202341070238 2023-10-16

Publications (1)

Publication Number Publication Date
WO2025083710A1 true WO2025083710A1 (en) 2025-04-24

Family

ID=95447926

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2024/052076 Pending WO2025083710A1 (en) 2023-10-16 2024-10-16 An antiviral photosensitive composition and device for potentiating the same

Country Status (1)

Country Link
WO (1) WO2025083710A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9302116B2 (en) * 2007-11-06 2016-04-05 Duke University Non-invasive energy upconversion methods and systems for in-situ photobiomodulation
WO2023009901A2 (en) * 2021-07-30 2023-02-02 DIFALCO, Raymond, J. Composition comprising photosensitzer compound and activation thereof
WO2023173087A1 (en) * 2022-03-11 2023-09-14 University Of Maryland, College Park Light-activatable, sustained-exposure ethanol injection
DE202024100465U1 (en) * 2024-01-31 2024-02-16 Siemens Healthineers Ag Medical device with several light strips with light-emitting diodes

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9302116B2 (en) * 2007-11-06 2016-04-05 Duke University Non-invasive energy upconversion methods and systems for in-situ photobiomodulation
WO2023009901A2 (en) * 2021-07-30 2023-02-02 DIFALCO, Raymond, J. Composition comprising photosensitzer compound and activation thereof
WO2023173087A1 (en) * 2022-03-11 2023-09-14 University Of Maryland, College Park Light-activatable, sustained-exposure ethanol injection
DE202024100465U1 (en) * 2024-01-31 2024-02-16 Siemens Healthineers Ag Medical device with several light strips with light-emitting diodes

Similar Documents

Publication Publication Date Title
Lei et al. Neuroprotective effects of quercetin in a mouse model of brain ischemic/reperfusion injury via anti-apoptotic mechanisms based on the Akt pathway
TWI667044B (en) Subcutaneous injection for reducing body weight and use thereof
EP2429498A4 (en) New oral formulations for tetrapyrrole derivatives
Zhao et al. Combination of metformin and paclitaxel suppresses proliferation and induces apoptosis of human prostate cancer cells via oxidative stress and targeting the mitochondria-dependent pathway
EP3193875B1 (en) Levocetirizine and montelukast in the treatment of inflammation mediated conditions
Fonseca-Berzal et al. Novel solid dispersions of benznidazole: preparation, dissolution profile and biological evaluation as alternative antichagasic drug delivery system
Pedrazini et al. L-arginine: Its role in human physiology, in some diseases and mainly in viral multiplication as a narrative literature review
US20120244212A1 (en) Enhanced method and composition for the treatment of hiv+ tuberculosis patients with anti-retroviral drugs and liposomal encapsulation for delivery of reduced glutathione
Chen et al. Kangfuxin promotes apoptosis of gastric cancer cells through activating ER-stress and autophagy
Wu et al. Pancreatic cancer cell apoptosis is induced by a proteoglycan extracted from Ganoderma lucidum
Chen et al. Antitumor effects and mechanisms of pyropheophorbide-α methyl ester-mediated photodynamic therapy on the human osteosarcoma cell line MG-63
Chen et al. Artesunate synergistically promotes sorafenib‑induced apoptosis and ferroptosis in non‑Hodgkin lymphoma cells through inhibition of the STAT3 pathway
Hu et al. Berberine inhibits excessive autophagy and protects myocardium against ischemia/reperfusion injury via the RhoE/AMPK pathway
CN113384698B (en) Self-assembled nano-medicament for synergetic chemotherapy/acousto-photodynamic therapy and application thereof
WO2020199460A1 (en) Use of luteolin-7-o-glucoside or luteolin-7-o-glucuronide in preparation of medicine for eye injuries
Rong et al. Methotrexate remediates spinal cord injury in vivo and in vitro via suppression of endoplasmic reticulum stress-induced apoptosis
WO2025083710A1 (en) An antiviral photosensitive composition and device for potentiating the same
CN103349776A (en) Edaravone injection solution and preparation method thereof
Amato et al. Treatment of mucosal leishmaniasis with a lipid formulation of amphotericin B
Wu et al. Effect of endoplasmic reticulum stress involved in manganese‑induced neurotoxicity in rats
WO2019045989A1 (en) Methods and compositions for the antiviral use of synthetic lysine analogs and mimetics
CN105796538A (en) Application of resveratrol and analogs of resveratrol in serving as HIV latent virus reservoir activating agents
US20210299077A1 (en) Liposomal reduced glutathione (lrg) in combination with ivermectin for the treatment of covid-19
BR112021001328A2 (en) lycopene compositions and methods to protect the skin from ultraviolet radiation
Lamer et al. Adverse cutaneous reactions to psychopharmaceuticals

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24879325

Country of ref document: EP

Kind code of ref document: A1