mRNA vaccine technology &
its applications beyond COVID-19
Dr Adarsh S
3rd Year PG student
Dept. of Microbiology
ESIC MC & PGIMSR
Rajajinagar, Bangalore
References
Al Fayez, N.; Nassar, M.S.; Alshehri, A.A.; Alnefaie, M.K.; Almughem, F.A.; Alshehri, B.Y.; Alawad, A.O.; Tawfik,
E.A. Recent Advancement in mRNA Vaccine Development and Applications. Pharmaceutics 2023, 15, 1972.
Wang YS, Kumari M, Chen GH, Hong MH, Yuan JP, Tsai JL, Wu HC. mRNA-based vaccines and therapeutics: an
in-depth survey of current and upcoming clinical applications. J Biomed Sci. 2023 Oct 7;30(1):84.
Chandra S, Wilson JC, Good D, Wei MQ. mRNA vaccines: a new era in vaccine development. Oncol Res. 2024 Sep
18;32(10):1543-1564
Parhiz H, Atochina-Vasserman EN, Weissman D. mRNA-based therapeutics: looking beyond COVID-19 vaccines.
Lancet. 2024 Mar 23;403(10432):1192-1204
Chen B, Yang Y, Wang X, Yang W, Lu Y, Wang D, Zhuo E, Tang Y, Su J, Tang G, Shao S, Gu K. mRNA
vaccine development and applications: A special focus on tumors (Review). Int J Oncol. 2024
Aug;65(2):81.
Ankrah PK, Ilesanmi A, Akinyemi AO, Lasehinde V, Adurosakin OE, Ajayi OH. Clinical Analysis and
Applications of mRNA Vaccines in Infectious Diseases and Cancer Treatment. Cureus. 2023 Oct
2;15(10):e46354.
Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug
Discov. 2018 Apr;17(4):261-279.
You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA
Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev. 2023 Mar
23;36(1)
Layout
 Central Dogma of Molecular Biology
 mRNA vaccine
 Advantages of mRNA Vaccines
 Disadvantages of mRNA Vaccines
 Types of mRNA Vaccines
 Mechanism of Action of mRNA Vaccines
 The mRNA Vaccine Delivery Systems
Polymeric Nanoparticles
Peptides and Proteins Nanoparticles
Protamine Nanoparticles
Lipid Nanoparticles (LNPs)
Exosome-based systems
 The mRNA Vaccine Applications
Infectious Disease Vaccines
Autoimmune & Rare Disease Therapies
Cancer Immunotherapy
Protein Replacement Therapy
Regenerative Medicine
Neurodegenerative Disease Therapies
Pandemic Preparedness & Biodefense
Gene Editing & CRISPR Applications
Purines
Pyramidines
Central Dogma of Molecular Biology
An mRNA vaccine is a type of vaccine that uses messenger RNA (mRNA) to instruct cells to produce a
specific antigenic protein, triggering an immune response without using a live pathogen.
Open Reading Frame
Katalin Karikó and Drew Weissman
Mechanism of Action of mRNA Vaccines
A conventional vaccine uses whole or parts of a pathogen (virus or bacteria) to stimulate the immune system and
provide immunity against infectious diseases.
Types
1.Live Attenuated Vaccines Use a weakened
form of the pathogen that can replicate but
does not cause disease.
MMR (Measles, Mumps, Rubella), BCG
(Tuberculosis), Oral Polio Vaccine (OPV).
2.Inactivated (Killed) Vaccines – Contain
whole pathogens that have been killed or
inactivated.
Inactivated Polio Vaccine (IPV), Hepatitis A
vaccine.
3. Subunit, Recombinant, or Conjugate Vaccines – Contain only
specific parts (antigens) of a pathogen.
Hepatitis B vaccine, HPV vaccine, Pneumococcal vaccine.
4. Toxoid Vaccines – Use inactivated bacterial toxins to generate
immunity. Tetanus and Diphtheria vaccines.
Advantages of mRNA Vaccines Over Conventional Vaccines
1. Faster Development and Deployment
2. No Need for Adjuvants
3. Safer for Immunocompromised Individuals
4. Stronger and More Balanced Immune Response
5. No Anti-Vector Immunity Issues
6. More Precise Antigen Expression
7. Reduced Risk of Allergic Reactions and Egg-Based
Contaminants
8. No Need for a Live Pathogen
9. No Risk of Genomic Integration
10. Easier Modification and Adaptability
11. Scalable and Cost-Effective Production
Disadvantages of mRNA Vaccines
1. Cold Storage and Stability Issues
2. Short mRNA Half-Life
3. Potential for Stronger Side Effects
4. Limited Long-Term Data
5. Lower Stability Compared to Conventional
Vaccines
6. Potential for Unintended Immune Reactions
7. Risk of Autoimmune Reactions
8. Manufacturing and Scalability Challenges
9. Limited Research on Non-Infectious Diseases
10. Need for Booster Doses
Types of mRNA Vaccines
mRNA vaccines can be classified into three main types based on their mechanism of action and ability to amplify
mRNA within cells.
1. Conventional mRNA Vaccines (Non-Replicating mRNA Vaccines)
Mechanism:
•Contains a single-stranded mRNA that encodes the antigen (e.g., viral spike protein).
•After delivery into cells, the mRNA is translated by ribosomes into the antigenic protein.
•The antigen is then presented to the immune system, triggering an immune response.
•The mRNA does not replicate and is eventually degraded by cellular processes.
Features:
 Simple design with no need for additional replication mechanisms.
 Requires higher doses because the mRNA does not replicate itself.
 Easier to manufacture and widely used in commercial vaccines.
Pfizer-BioNTech and Moderna COVID-19 vaccines.
2. Self-Amplifying mRNA (saRNA) Vaccines (Replicon mRNA Vaccines)
Mechanism:
•Contains two components:
• mRNA encoding the antigen (target protein).
• mRNA encoding viral replication machinery (usually from alphaviruses, like RNA-dependent RNA polymerase).
•Once inside the cell, the viral replication machinery amplifies the mRNA, producing multiple copies of the antigenic protein.
•This increases antigen production and enhances immune response with a lower initial mRNA dose.
Features:
✔ Higher efficiency -requires a lower dose than conventional mRNA vaccines.
✔ Longer-lasting antigen production, leading to a stronger immune response.
✔ Still in experimental stages – under development for flu, Zika, and cancer vaccines.
Example: Experimental saRNA COVID-19 vaccines by Imperial College London and Gritstone Bio.
3. Trans-Amplifying mRNA (taRNA) Vaccines
Mechanism:
•Uses two separate mRNA molecules:
• Antigen-encoding mRNA (like in conventional vaccines).
• Helper mRNA that provides replication enzymes (but not the full viral machinery).
•Unlike self-amplifying mRNA, where the replication genes are on the same strand as the antigen, trans-amplifying
mRNA vaccines separate them, allowing flexibility in vaccine design.
Features:
 More controlled replication than self-amplifying mRNA, reducing unwanted side effects.
 Lower risk of immune responses against viral replication proteins, which is a concern in saRNA vaccines.
 Still in early research stages, with potential applications in infectious diseases and cancer therapy
The mRNA Vaccine Delivery Systems
Polymeric Nanoparticles
Peptides and Proteins Nanoparticles
Protamine Nanoparticles
Lipid Nanoparticles (LNPs)
Exosome-based systems
Polymeric Nanoparticles
Encapsulation: mRNA binds to cationic polymers for protection.
Cellular Uptake: Nanoparticles enter cells via endocytosis.
Endosomal Escape: pH-responsive polymers release mRNA into the cytoplasm.
Degradation: The polymeric carrier is broken down and cleared.
Targeting ligands can be
added to the polymeric
surface for cell-specific
delivery (e.g., targeting
dendritic cells or tumor
cells).
• Polyethyleneimine (PEI)
– Effective but can be
toxic.
• Poly(β-amino esters)
(PBAEs) – More
biodegradable and less
toxic than PEI.
• Chitosan Nanoparticle
Peptides and Proteins Nanoparticles
mRNA Encapsulation & Protection – Cationic
peptides (e.g., arginine-rich peptides) electrostatically
bind to negatively charged mRNA, forming stable
nano-complexes that protect mRNA from degradation.
Cellular Uptake (Endocytosis & Membrane
Fusion) – Peptide-mRNA complexes enter cells via
endocytosis, and pH-sensitive cell-penetrating
peptides (CPPs) like RALA peptides alter their
structure in acidic endosomes, facilitating membrane
fusion.
Endosomal Escape & mRNA Release – Peptides
disrupt endosomal membranes, allowing mRNA
release into the cytoplasm, preventing degradation and
enabling translation
repeated
arginine-
alanine-
leucine-
alanine
Protamine Nanoparticles
mRNA Encapsulation & Protection – Protamine, a cationic protein rich in arginine, electrostatically binds to
negatively charged mRNA, forming stable nano-complexes..
Cellular Uptake (Endocytosis) – The protamine-mRNA complex enters the cell via endocytosis.
Endosomal Escape & Cytoplasmic Release – Protamine disrupts endosomes, releasing mRNA into the cytoplasm
Lipid Nanoparticles (LNPs)
Encapsulation & Protection – LNPs shield mRNA from degradation.
Cellular Uptake – LNPs enter cells via endocytosis.
Endosomal Escape – Ionizable lipids disrupt endosomes, releasing mRNA into the cytoplasm.
Translation & Immune Activation – mRNA is translated into an antigen, triggering an immune response.
LNPs typically consist of:
✔ Ionizable lipids (40–50%) – Aid in mRNA
binding and endosomal escape.
✔ Cholesterol (38–45%) – Enhances membrane
stability.
✔ Helper phospholipids (10–12%) – Assist in
lipid bilayer fusion.
✔ PEGylated lipids (1–2%) – Improve circulation
time and prevent aggregation.
Microfluidics Process for LNP-mRNA Formulation
Controlled Mixing – mRNA (aqueous buffer) and lipids (ethanol) are precisely mixed in a microfluidic device.
Self-Assembly of LNPs – Lipids encapsulate mRNA, forming stable LNP-mRNA complexes with high efficiency.
Size Control & Homogeneous Formation – Microfluidic channels regulate LNP size (~50-100 nm) for consistent delivery.
Purification (Tangential Flow Filtration - TFF) – Removes excess ethanol and buffers, stabilizing the formulation.
Sterilisation & Final Product – The purified LNP-mRNA is filtered (0.2 μm) and prepared in liquid or lyophilized form
Encapsulation & Engineering
Cellular Uptake
Endosomal Escape
Translation & Immune Activation
Exosome-based systems
small extracellular vesicles (EVs) (30–150 nm) secreted by
cells that play a crucial role in cell-to-cell communication by
transporting proteins, lipids, RNA, and mRNA between cells.
They originate from endosomal compartments and are
released when multivesicular bodies (MVBs) fuse with the
plasma membrane
The mRNA Vaccine Applications
First mRNA therapy
was tested in 1992
(vasopressin-
encoding mRNA for
diabetes insipidus)
Infectious Disease Vaccines
Cancer Immunotherapy
Autoimmune & Rare Disease Therapies
Protein Replacement Therapy
Regenerative Medicine
Gene Editing & CRISPR Applications
Neurodegenerative Disease Therapies
Pandemic Preparedness & Biodefense
Infectious Disease Vaccines
Current mRNA Vaccines (Approved & In Use)
1.COVID-19 (SARS-CoV-2)
1. Pfizer-BioNTech (BNT162b2) & Moderna (mRNA-1273)
2. First-ever mRNA vaccines approved.
2.Influenza (Clinical trials – Phase 3)
1. Moderna’s mRNA-1010, targeting multiple flu strains.
Future mRNA Vaccines (Under Development)
3.Zika Virus – Preclinical & early-phase trials for high-risk areas.
4.Rabies – CureVac’s CV7202 shows promising immunogenicity.
5.Cytomegalovirus (CMV) – Moderna’s mRNA-1647 in clinical trials.
6.HIV – mRNA-1644 (Moderna & NIH) in Phase 1 trials.
7.Tuberculosis (TB) – Potential mRNA alternative to BCG vaccine.
8.Malaria – Targeting Plasmodium species, under development.
9.Dengue, Chikungunya, Ebola – Various early-stage trials.
Autoimmune & Rare Disease Therapies
1. Autoimmune Diseases
•Multiple Sclerosis (MS): mRNA therapy trains immune tolerance to myelin antigens.
•Type 1 Diabetes: mRNA helps protect and regenerate pancreatic beta cells.
•Inflammatory Bowel Disease (IBD): mRNA modulates immune response to reduce gut
inflammation.
2. Rare Genetic Disorders
•Cystic Fibrosis (CF): mRNA therapy restores CFTR protein function.
•Propionic Acidemia & Methylmalonic Acidemia: Corrects enzyme deficiencies.
•Lysosomal Storage Disorders (e.g., Gaucher, Pompe, Fabry disease): mRNA replaces
missing or defective enzymes.
•Glycogen Storage Diseases: mRNA restores glycogen metabolism enzymes.
(Ongoing Research & Clinical Trials)
reprogramming the
immune system to restore
immune tolerance and
prevent excessive immune
responses.
introducing specific
mRNA sequences that
encode tolerogenic
proteins or anti-
inflammatory factors
to regulate the
immune response.
Future Applications (Emerging Research & Potential Uses)
Autoimmune & Rare Disease Therapies
Personalized Autoimmune Therapies
•Tailored mRNA vaccines to modulate immune response for Rheumatoid Arthritis, Lupus, IBD and Psoriasis.
Tolerogenic mRNA Vaccines
•Teaches the immune system to recognize self-antigens, preventing autoimmune attacks.
•Potential in Multiple Sclerosis, Type 1 Diabetes, Myasthenia Gravis and Autoimmune Hepatitis.
Gene Editing & Protein Replacement Therapies
•Duchenne Muscular Dystrophy (DMD): mRNA increases dystrophin protein expression.
•Hemophilia: mRNA-based clotting factor replacement.
Cancer Immunotherapy
Personalized Cancer Vaccines
•mRNA-based neoantigen vaccines stimulate immune responses against patient-specific tumor mutations.
(Moderna & BioNTech’s mRNA cancer vaccines in clinical trials for melanoma, lung, and pancreatic cancer)
Off-the-Shelf Cancer Vaccines
•Targets common tumor-associated antigens (TAAs) like HER2 (breast cancer), MAGE-A3 (lung cancer), NY-ESO-1
(sarcoma & melanoma).
mRNA-Encoded Cytokine Therapy
• mRNA delivers cytokines (e.g., IL-2, IL-12, IFN-γ) to enhance anti-tumor immune response.
• Helps overcome immunosuppressive tumor microenvironments.
mRNA for Chimeric Antigen Receptor (CAR) T & NK Cell Therapy
• mRNA engineering of CAR-T and CAR-NK cells enhances their ability to target cancers.
• Avoids permanent genetic modifications, improving safety.
Cancer Immunotherapy
Protein Replacement Therapy
Monoclonal Antibody Production
mRNA instructs cells to produce therapeutic
monoclonal antibodies inside the body, reducing
the need for external antibody infusions.
Hormone Replacement Therapy
Insulin (Diabetes): mRNA-based insulin therapy
to restore glucose regulation.
Growth Hormone Deficiency: mRNA therapy for
controlled human growth hormone (hGH)
production.
Clotting Factor Therapy
(Hemophilia)
mRNA encodes Factor VIII or IX to
treat Hemophilia A and B, reducing
dependency on regular injections.
Erythropoietin (EPO) Therapy
(Anemia & Kidney Disease)
mRNA-based EPO production for
chronic kidney disease and
chemotherapy-induced anemia.
Protein Replacement Therapy
Lysosomal Storage Disease Therapy
mRNA-based replacement of missing
enzymes for Gaucher, Fabry, Pompe,
and Tay-Sachs diseases.
Cystic Fibrosis (CF) Therapy
mRNA restores CFTR protein function,
addressing the root cause of CF.
mRNA-Based Therapies for
Muscular Dystrophies
Duchenne Muscular Dystrophy (DMD):
mRNA therapy enhances dystrophin
production.
Enzyme Replacement for
Metabolic Disorders
mRNA encodes metabolic enzymes
for disorders like Phenylketonuria
(PKU) and Glycogen Storage
Diseases.
mRNA for Neurodegenerative
Disease Treatments
Alzheimer’s & Parkinson’s Disease:
mRNA restores neuroprotective
proteins to slow disease progression.
Self-Amplifying mRNA for Long-
Lasting Protein Therapy
Regenerative Medicine
• mRNA-based therapy to promote cardiac muscle repair after heart attacks
(myocardial infarction).
• Encodes growth factors like VEGF (vascular endothelial growth factor) to
stimulate new blood vessel formation.
Cardiac Regeneration (Heart
Disease Treatment)
• mRNA instructs cells to produce collagen and growth factors to accelerate
wound closure and tissue healing.
• Potential for burn treatment and diabetic wound repair.
Wound Healing & Skin
Regeneration
• mRNA delivers bone morphogenetic proteins (BMPs) to enhance bone and
cartilage repair.
• Studied for treating osteoporosis, arthritis, and non-healing fractures.
Cartilage & Bone
Regeneration (Osteoarthritis
& Fracture Healing)
• mRNA therapy to stimulate neural regeneration and promote functional recovery
in spinal cord injury patients.
Spinal Cord Injury Repair
• mRNA-driven therapies to repair liver damage caused by cirrhosis, liver failure,
or chronic diseases.
Liver Regeneration
• mRNA-encoded neurotrophic factors to regenerate neurons in
neurodegenerative diseases like Alzheimer’s, Parkinson’s, and stroke recovery.
Neuroregeneration (Brain & Nerve
Repair)
• mRNA instructs cells to regenerate insulin-producing beta cells, offering a
potential cure for Type 1 Diabetes.
Regeneration of Pancreatic Beta
Cells (Diabetes Treatment)
• mRNA encodes proteins to repair damaged lung tissue in chronic obstructive
pulmonary disease (COPD) and pulmonary fibrosis.
Lung Regeneration (Respiratory
Diseases)
• Converts adult cells into pluripotent stem cells (iPSCs) for personalized
regenerative medicine.
mRNA-Induced Stem Cell Therapy
Self-Amplifying mRNA (saRNA) for Long-
Term Regeneration
Neurodegenerative Disease Therapies
Alzheimer’s Disease (AD)
mRNA therapy encodes neuroprotective proteins (e.g., brain-derived neurotrophic
factor BDNF) to support neuron survival.
mRNA-based beta-amyloid and tau-targeting antibodies to slow disease
progression.
Parkinson’s Disease (PD)
mRNA therapy stimulates dopamine-producing neurons by delivering growth
factors like glial cell line-derived neurotrophic factor (GDNF).
Amyotrophic Lateral Sclerosis (ALS)
mRNA-based therapy targets superoxide dismutase 1 (SOD1) mutations, a major cause of ALS.
Encourages motor neuron survival and function.
Huntington’s Disease (HD)
mRNA therapy designed to reduce mutant huntingtin protein (mHTT) levels, preventing neuronal damage
mRNA for Stroke Recovery & Neural Regeneration
mRNA-driven therapies to stimulate angiogenesis and neurogenesis after ischemic stroke.
• Allows lower-dose vaccines with prolonged protection, ideal for mass
immunization during outbreaks.
Self-Amplifying mRNA (saRNA) for
Pandemic Response
• Developing mRNA-based countermeasures for anthrax, smallpox,
and hemorrhagic fever viruses (Ebola, Marburg, Lassa fever).
mRNA Vaccines for Bioterrorism
Threats
• WHO and global research groups using AI-driven mRNA design to
predict and prepare for unknown future pathogens.
mRNA Vaccines for High-Risk Viruses
("Disease X")
• Next-gen mRNA vaccines stored at room temperature, solving cold-
chain logistics issues.
Thermostable mRNA Vaccines for
Global Readiness
• On-demand mRNA therapies producing neutralizing antibodies
against new viral threats.
mRNA-Based Monoclonal Antibodies
for Rapid Defense
Combination mRNA Vaccines for
Multi-Pathogen Protection
Single-dose vaccines covering multiple
viruses (e.g., COVID-19 + Flu + RSV).
Pandemic Preparedness & Biodefense
mRNA vaccine technology in India
Gennova Biopharmaceuticals Ltd. (Pune)
•Developed GEMCOVAC®-19, India's first mRNA COVID-19 vaccine.
•Created GEMCOVAC®-OM, an Omicron-specific booster.
Serum Institute of India (SII) (Pune)
•Exploring mRNA-based cancer immunotherapy in collaboration with
Univercells (Belgium).
Centre for Cellular and Molecular Biology (CCMB) (Hyderabad)
•Developed India’s first indigenous mRNA vaccine technology with
CSIR
mRNA is no longer just about vaccines—it’s the future of medicine!
mRNA vaccine technology and its applications, beyond COVID-19. Dr Adarsh Soman

mRNA vaccine technology and its applications, beyond COVID-19. Dr Adarsh Soman

  • 1.
    mRNA vaccine technology& its applications beyond COVID-19 Dr Adarsh S 3rd Year PG student Dept. of Microbiology ESIC MC & PGIMSR Rajajinagar, Bangalore
  • 2.
    References Al Fayez, N.;Nassar, M.S.; Alshehri, A.A.; Alnefaie, M.K.; Almughem, F.A.; Alshehri, B.Y.; Alawad, A.O.; Tawfik, E.A. Recent Advancement in mRNA Vaccine Development and Applications. Pharmaceutics 2023, 15, 1972. Wang YS, Kumari M, Chen GH, Hong MH, Yuan JP, Tsai JL, Wu HC. mRNA-based vaccines and therapeutics: an in-depth survey of current and upcoming clinical applications. J Biomed Sci. 2023 Oct 7;30(1):84. Chandra S, Wilson JC, Good D, Wei MQ. mRNA vaccines: a new era in vaccine development. Oncol Res. 2024 Sep 18;32(10):1543-1564 Parhiz H, Atochina-Vasserman EN, Weissman D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet. 2024 Mar 23;403(10432):1192-1204
  • 3.
    Chen B, YangY, Wang X, Yang W, Lu Y, Wang D, Zhuo E, Tang Y, Su J, Tang G, Shao S, Gu K. mRNA vaccine development and applications: A special focus on tumors (Review). Int J Oncol. 2024 Aug;65(2):81. Ankrah PK, Ilesanmi A, Akinyemi AO, Lasehinde V, Adurosakin OE, Ajayi OH. Clinical Analysis and Applications of mRNA Vaccines in Infectious Diseases and Cancer Treatment. Cureus. 2023 Oct 2;15(10):e46354. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018 Apr;17(4):261-279. You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev. 2023 Mar 23;36(1)
  • 4.
    Layout  Central Dogmaof Molecular Biology  mRNA vaccine  Advantages of mRNA Vaccines  Disadvantages of mRNA Vaccines  Types of mRNA Vaccines  Mechanism of Action of mRNA Vaccines  The mRNA Vaccine Delivery Systems Polymeric Nanoparticles Peptides and Proteins Nanoparticles Protamine Nanoparticles Lipid Nanoparticles (LNPs) Exosome-based systems  The mRNA Vaccine Applications Infectious Disease Vaccines Autoimmune & Rare Disease Therapies Cancer Immunotherapy Protein Replacement Therapy Regenerative Medicine Neurodegenerative Disease Therapies Pandemic Preparedness & Biodefense Gene Editing & CRISPR Applications
  • 5.
  • 6.
    Central Dogma ofMolecular Biology
  • 10.
    An mRNA vaccineis a type of vaccine that uses messenger RNA (mRNA) to instruct cells to produce a specific antigenic protein, triggering an immune response without using a live pathogen. Open Reading Frame
  • 11.
    Katalin Karikó andDrew Weissman
  • 12.
    Mechanism of Actionof mRNA Vaccines
  • 14.
    A conventional vaccineuses whole or parts of a pathogen (virus or bacteria) to stimulate the immune system and provide immunity against infectious diseases. Types 1.Live Attenuated Vaccines Use a weakened form of the pathogen that can replicate but does not cause disease. MMR (Measles, Mumps, Rubella), BCG (Tuberculosis), Oral Polio Vaccine (OPV). 2.Inactivated (Killed) Vaccines – Contain whole pathogens that have been killed or inactivated. Inactivated Polio Vaccine (IPV), Hepatitis A vaccine. 3. Subunit, Recombinant, or Conjugate Vaccines – Contain only specific parts (antigens) of a pathogen. Hepatitis B vaccine, HPV vaccine, Pneumococcal vaccine. 4. Toxoid Vaccines – Use inactivated bacterial toxins to generate immunity. Tetanus and Diphtheria vaccines.
  • 15.
    Advantages of mRNAVaccines Over Conventional Vaccines 1. Faster Development and Deployment 2. No Need for Adjuvants 3. Safer for Immunocompromised Individuals 4. Stronger and More Balanced Immune Response 5. No Anti-Vector Immunity Issues 6. More Precise Antigen Expression 7. Reduced Risk of Allergic Reactions and Egg-Based Contaminants 8. No Need for a Live Pathogen 9. No Risk of Genomic Integration 10. Easier Modification and Adaptability 11. Scalable and Cost-Effective Production Disadvantages of mRNA Vaccines 1. Cold Storage and Stability Issues 2. Short mRNA Half-Life 3. Potential for Stronger Side Effects 4. Limited Long-Term Data 5. Lower Stability Compared to Conventional Vaccines 6. Potential for Unintended Immune Reactions 7. Risk of Autoimmune Reactions 8. Manufacturing and Scalability Challenges 9. Limited Research on Non-Infectious Diseases 10. Need for Booster Doses
  • 16.
    Types of mRNAVaccines mRNA vaccines can be classified into three main types based on their mechanism of action and ability to amplify mRNA within cells. 1. Conventional mRNA Vaccines (Non-Replicating mRNA Vaccines) Mechanism: •Contains a single-stranded mRNA that encodes the antigen (e.g., viral spike protein). •After delivery into cells, the mRNA is translated by ribosomes into the antigenic protein. •The antigen is then presented to the immune system, triggering an immune response. •The mRNA does not replicate and is eventually degraded by cellular processes.
  • 17.
    Features:  Simple designwith no need for additional replication mechanisms.  Requires higher doses because the mRNA does not replicate itself.  Easier to manufacture and widely used in commercial vaccines. Pfizer-BioNTech and Moderna COVID-19 vaccines.
  • 18.
    2. Self-Amplifying mRNA(saRNA) Vaccines (Replicon mRNA Vaccines) Mechanism: •Contains two components: • mRNA encoding the antigen (target protein). • mRNA encoding viral replication machinery (usually from alphaviruses, like RNA-dependent RNA polymerase). •Once inside the cell, the viral replication machinery amplifies the mRNA, producing multiple copies of the antigenic protein. •This increases antigen production and enhances immune response with a lower initial mRNA dose. Features: ✔ Higher efficiency -requires a lower dose than conventional mRNA vaccines. ✔ Longer-lasting antigen production, leading to a stronger immune response. ✔ Still in experimental stages – under development for flu, Zika, and cancer vaccines. Example: Experimental saRNA COVID-19 vaccines by Imperial College London and Gritstone Bio.
  • 19.
    3. Trans-Amplifying mRNA(taRNA) Vaccines Mechanism: •Uses two separate mRNA molecules: • Antigen-encoding mRNA (like in conventional vaccines). • Helper mRNA that provides replication enzymes (but not the full viral machinery). •Unlike self-amplifying mRNA, where the replication genes are on the same strand as the antigen, trans-amplifying mRNA vaccines separate them, allowing flexibility in vaccine design. Features:  More controlled replication than self-amplifying mRNA, reducing unwanted side effects.  Lower risk of immune responses against viral replication proteins, which is a concern in saRNA vaccines.  Still in early research stages, with potential applications in infectious diseases and cancer therapy
  • 21.
    The mRNA VaccineDelivery Systems Polymeric Nanoparticles Peptides and Proteins Nanoparticles Protamine Nanoparticles Lipid Nanoparticles (LNPs) Exosome-based systems
  • 22.
    Polymeric Nanoparticles Encapsulation: mRNAbinds to cationic polymers for protection. Cellular Uptake: Nanoparticles enter cells via endocytosis. Endosomal Escape: pH-responsive polymers release mRNA into the cytoplasm. Degradation: The polymeric carrier is broken down and cleared. Targeting ligands can be added to the polymeric surface for cell-specific delivery (e.g., targeting dendritic cells or tumor cells). • Polyethyleneimine (PEI) – Effective but can be toxic. • Poly(β-amino esters) (PBAEs) – More biodegradable and less toxic than PEI. • Chitosan Nanoparticle
  • 23.
    Peptides and ProteinsNanoparticles mRNA Encapsulation & Protection – Cationic peptides (e.g., arginine-rich peptides) electrostatically bind to negatively charged mRNA, forming stable nano-complexes that protect mRNA from degradation. Cellular Uptake (Endocytosis & Membrane Fusion) – Peptide-mRNA complexes enter cells via endocytosis, and pH-sensitive cell-penetrating peptides (CPPs) like RALA peptides alter their structure in acidic endosomes, facilitating membrane fusion. Endosomal Escape & mRNA Release – Peptides disrupt endosomal membranes, allowing mRNA release into the cytoplasm, preventing degradation and enabling translation repeated arginine- alanine- leucine- alanine
  • 24.
    Protamine Nanoparticles mRNA Encapsulation& Protection – Protamine, a cationic protein rich in arginine, electrostatically binds to negatively charged mRNA, forming stable nano-complexes.. Cellular Uptake (Endocytosis) – The protamine-mRNA complex enters the cell via endocytosis. Endosomal Escape & Cytoplasmic Release – Protamine disrupts endosomes, releasing mRNA into the cytoplasm
  • 25.
    Lipid Nanoparticles (LNPs) Encapsulation& Protection – LNPs shield mRNA from degradation. Cellular Uptake – LNPs enter cells via endocytosis. Endosomal Escape – Ionizable lipids disrupt endosomes, releasing mRNA into the cytoplasm. Translation & Immune Activation – mRNA is translated into an antigen, triggering an immune response.
  • 26.
    LNPs typically consistof: ✔ Ionizable lipids (40–50%) – Aid in mRNA binding and endosomal escape. ✔ Cholesterol (38–45%) – Enhances membrane stability. ✔ Helper phospholipids (10–12%) – Assist in lipid bilayer fusion. ✔ PEGylated lipids (1–2%) – Improve circulation time and prevent aggregation.
  • 27.
    Microfluidics Process forLNP-mRNA Formulation Controlled Mixing – mRNA (aqueous buffer) and lipids (ethanol) are precisely mixed in a microfluidic device. Self-Assembly of LNPs – Lipids encapsulate mRNA, forming stable LNP-mRNA complexes with high efficiency. Size Control & Homogeneous Formation – Microfluidic channels regulate LNP size (~50-100 nm) for consistent delivery. Purification (Tangential Flow Filtration - TFF) – Removes excess ethanol and buffers, stabilizing the formulation. Sterilisation & Final Product – The purified LNP-mRNA is filtered (0.2 μm) and prepared in liquid or lyophilized form
  • 29.
    Encapsulation & Engineering CellularUptake Endosomal Escape Translation & Immune Activation Exosome-based systems small extracellular vesicles (EVs) (30–150 nm) secreted by cells that play a crucial role in cell-to-cell communication by transporting proteins, lipids, RNA, and mRNA between cells. They originate from endosomal compartments and are released when multivesicular bodies (MVBs) fuse with the plasma membrane
  • 30.
    The mRNA VaccineApplications First mRNA therapy was tested in 1992 (vasopressin- encoding mRNA for diabetes insipidus) Infectious Disease Vaccines Cancer Immunotherapy Autoimmune & Rare Disease Therapies Protein Replacement Therapy Regenerative Medicine Gene Editing & CRISPR Applications Neurodegenerative Disease Therapies Pandemic Preparedness & Biodefense
  • 31.
    Infectious Disease Vaccines CurrentmRNA Vaccines (Approved & In Use) 1.COVID-19 (SARS-CoV-2) 1. Pfizer-BioNTech (BNT162b2) & Moderna (mRNA-1273) 2. First-ever mRNA vaccines approved. 2.Influenza (Clinical trials – Phase 3) 1. Moderna’s mRNA-1010, targeting multiple flu strains. Future mRNA Vaccines (Under Development) 3.Zika Virus – Preclinical & early-phase trials for high-risk areas. 4.Rabies – CureVac’s CV7202 shows promising immunogenicity. 5.Cytomegalovirus (CMV) – Moderna’s mRNA-1647 in clinical trials. 6.HIV – mRNA-1644 (Moderna & NIH) in Phase 1 trials. 7.Tuberculosis (TB) – Potential mRNA alternative to BCG vaccine. 8.Malaria – Targeting Plasmodium species, under development. 9.Dengue, Chikungunya, Ebola – Various early-stage trials.
  • 32.
    Autoimmune & RareDisease Therapies 1. Autoimmune Diseases •Multiple Sclerosis (MS): mRNA therapy trains immune tolerance to myelin antigens. •Type 1 Diabetes: mRNA helps protect and regenerate pancreatic beta cells. •Inflammatory Bowel Disease (IBD): mRNA modulates immune response to reduce gut inflammation. 2. Rare Genetic Disorders •Cystic Fibrosis (CF): mRNA therapy restores CFTR protein function. •Propionic Acidemia & Methylmalonic Acidemia: Corrects enzyme deficiencies. •Lysosomal Storage Disorders (e.g., Gaucher, Pompe, Fabry disease): mRNA replaces missing or defective enzymes. •Glycogen Storage Diseases: mRNA restores glycogen metabolism enzymes. (Ongoing Research & Clinical Trials) reprogramming the immune system to restore immune tolerance and prevent excessive immune responses. introducing specific mRNA sequences that encode tolerogenic proteins or anti- inflammatory factors to regulate the immune response.
  • 33.
    Future Applications (EmergingResearch & Potential Uses) Autoimmune & Rare Disease Therapies Personalized Autoimmune Therapies •Tailored mRNA vaccines to modulate immune response for Rheumatoid Arthritis, Lupus, IBD and Psoriasis. Tolerogenic mRNA Vaccines •Teaches the immune system to recognize self-antigens, preventing autoimmune attacks. •Potential in Multiple Sclerosis, Type 1 Diabetes, Myasthenia Gravis and Autoimmune Hepatitis. Gene Editing & Protein Replacement Therapies •Duchenne Muscular Dystrophy (DMD): mRNA increases dystrophin protein expression. •Hemophilia: mRNA-based clotting factor replacement.
  • 34.
    Cancer Immunotherapy Personalized CancerVaccines •mRNA-based neoantigen vaccines stimulate immune responses against patient-specific tumor mutations. (Moderna & BioNTech’s mRNA cancer vaccines in clinical trials for melanoma, lung, and pancreatic cancer) Off-the-Shelf Cancer Vaccines •Targets common tumor-associated antigens (TAAs) like HER2 (breast cancer), MAGE-A3 (lung cancer), NY-ESO-1 (sarcoma & melanoma).
  • 35.
    mRNA-Encoded Cytokine Therapy •mRNA delivers cytokines (e.g., IL-2, IL-12, IFN-γ) to enhance anti-tumor immune response. • Helps overcome immunosuppressive tumor microenvironments. mRNA for Chimeric Antigen Receptor (CAR) T & NK Cell Therapy • mRNA engineering of CAR-T and CAR-NK cells enhances their ability to target cancers. • Avoids permanent genetic modifications, improving safety. Cancer Immunotherapy
  • 36.
    Protein Replacement Therapy MonoclonalAntibody Production mRNA instructs cells to produce therapeutic monoclonal antibodies inside the body, reducing the need for external antibody infusions. Hormone Replacement Therapy Insulin (Diabetes): mRNA-based insulin therapy to restore glucose regulation. Growth Hormone Deficiency: mRNA therapy for controlled human growth hormone (hGH) production. Clotting Factor Therapy (Hemophilia) mRNA encodes Factor VIII or IX to treat Hemophilia A and B, reducing dependency on regular injections. Erythropoietin (EPO) Therapy (Anemia & Kidney Disease) mRNA-based EPO production for chronic kidney disease and chemotherapy-induced anemia.
  • 37.
    Protein Replacement Therapy LysosomalStorage Disease Therapy mRNA-based replacement of missing enzymes for Gaucher, Fabry, Pompe, and Tay-Sachs diseases. Cystic Fibrosis (CF) Therapy mRNA restores CFTR protein function, addressing the root cause of CF. mRNA-Based Therapies for Muscular Dystrophies Duchenne Muscular Dystrophy (DMD): mRNA therapy enhances dystrophin production. Enzyme Replacement for Metabolic Disorders mRNA encodes metabolic enzymes for disorders like Phenylketonuria (PKU) and Glycogen Storage Diseases. mRNA for Neurodegenerative Disease Treatments Alzheimer’s & Parkinson’s Disease: mRNA restores neuroprotective proteins to slow disease progression. Self-Amplifying mRNA for Long- Lasting Protein Therapy
  • 38.
    Regenerative Medicine • mRNA-basedtherapy to promote cardiac muscle repair after heart attacks (myocardial infarction). • Encodes growth factors like VEGF (vascular endothelial growth factor) to stimulate new blood vessel formation. Cardiac Regeneration (Heart Disease Treatment) • mRNA instructs cells to produce collagen and growth factors to accelerate wound closure and tissue healing. • Potential for burn treatment and diabetic wound repair. Wound Healing & Skin Regeneration • mRNA delivers bone morphogenetic proteins (BMPs) to enhance bone and cartilage repair. • Studied for treating osteoporosis, arthritis, and non-healing fractures. Cartilage & Bone Regeneration (Osteoarthritis & Fracture Healing)
  • 39.
    • mRNA therapyto stimulate neural regeneration and promote functional recovery in spinal cord injury patients. Spinal Cord Injury Repair • mRNA-driven therapies to repair liver damage caused by cirrhosis, liver failure, or chronic diseases. Liver Regeneration • mRNA-encoded neurotrophic factors to regenerate neurons in neurodegenerative diseases like Alzheimer’s, Parkinson’s, and stroke recovery. Neuroregeneration (Brain & Nerve Repair) • mRNA instructs cells to regenerate insulin-producing beta cells, offering a potential cure for Type 1 Diabetes. Regeneration of Pancreatic Beta Cells (Diabetes Treatment) • mRNA encodes proteins to repair damaged lung tissue in chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. Lung Regeneration (Respiratory Diseases) • Converts adult cells into pluripotent stem cells (iPSCs) for personalized regenerative medicine. mRNA-Induced Stem Cell Therapy Self-Amplifying mRNA (saRNA) for Long- Term Regeneration
  • 40.
    Neurodegenerative Disease Therapies Alzheimer’sDisease (AD) mRNA therapy encodes neuroprotective proteins (e.g., brain-derived neurotrophic factor BDNF) to support neuron survival. mRNA-based beta-amyloid and tau-targeting antibodies to slow disease progression. Parkinson’s Disease (PD) mRNA therapy stimulates dopamine-producing neurons by delivering growth factors like glial cell line-derived neurotrophic factor (GDNF).
  • 41.
    Amyotrophic Lateral Sclerosis(ALS) mRNA-based therapy targets superoxide dismutase 1 (SOD1) mutations, a major cause of ALS. Encourages motor neuron survival and function. Huntington’s Disease (HD) mRNA therapy designed to reduce mutant huntingtin protein (mHTT) levels, preventing neuronal damage mRNA for Stroke Recovery & Neural Regeneration mRNA-driven therapies to stimulate angiogenesis and neurogenesis after ischemic stroke.
  • 42.
    • Allows lower-dosevaccines with prolonged protection, ideal for mass immunization during outbreaks. Self-Amplifying mRNA (saRNA) for Pandemic Response • Developing mRNA-based countermeasures for anthrax, smallpox, and hemorrhagic fever viruses (Ebola, Marburg, Lassa fever). mRNA Vaccines for Bioterrorism Threats • WHO and global research groups using AI-driven mRNA design to predict and prepare for unknown future pathogens. mRNA Vaccines for High-Risk Viruses ("Disease X") • Next-gen mRNA vaccines stored at room temperature, solving cold- chain logistics issues. Thermostable mRNA Vaccines for Global Readiness • On-demand mRNA therapies producing neutralizing antibodies against new viral threats. mRNA-Based Monoclonal Antibodies for Rapid Defense Combination mRNA Vaccines for Multi-Pathogen Protection Single-dose vaccines covering multiple viruses (e.g., COVID-19 + Flu + RSV). Pandemic Preparedness & Biodefense
  • 43.
    mRNA vaccine technologyin India Gennova Biopharmaceuticals Ltd. (Pune) •Developed GEMCOVAC®-19, India's first mRNA COVID-19 vaccine. •Created GEMCOVAC®-OM, an Omicron-specific booster. Serum Institute of India (SII) (Pune) •Exploring mRNA-based cancer immunotherapy in collaboration with Univercells (Belgium). Centre for Cellular and Molecular Biology (CCMB) (Hyderabad) •Developed India’s first indigenous mRNA vaccine technology with CSIR
  • 46.
    mRNA is nolonger just about vaccines—it’s the future of medicine!

Editor's Notes

  • #5 Ribonucleic acid (RNA) is a polymer of purine and pyrimidine nucleotides linked by phosphodiester bonds. Messenger RNA (mRNA). Ribosomal RNA (rRNA). Transfer RNA (tRNA). Small RNA Micro-RNA (miRNA).
  • #6 the information available in the DNA is passed to messenger RNA, which is then used for synthesis of a particular protein DNA replication is like printing a copy of a book’s pages. The replication process occurs only at the time of cell division. But transcription is taking place all the time. Only certain areas of the DNA are copied (selected regions on the sense strand). This is like taking a Xerox copy of a particular page of the book. So, the genetic information of DNA is transcribed (copied) to the messenger RNA (mRNA). During transcription, the message from the DNA is copied in the language of nucleotides (4-letter language). The mRNA then reaches the cytoplasm, which is translated into functional proteins. During translation, the nucleotide sequence is translated to the language of amino acid sequence
  • #7 The mRNA base sequence is complementary to that of the template strand and identical to that of the coding strand. In mRNA, U replaces T It acts as a messenger of the information in the gene in DNA to the protein-synthesizing machinery in the cytoplasm. It carries the message to be translated to a protein. The template strand of DNA is transcribed into a single-stranded mRNA. This is accomplished by the DNA-dependent RNA polymerase. The mRNA is a complementary copy of the template strand of the DNA. However, thymine is not present in RNA; instead, uracil will be incorporated
  • #10 The immune system recognizes this antigen as foreign, activating T-cell and B-cell responses to generate immunity. The mRNA does not integrate into the genome and is eventually degraded after protein translation.
  • #11 Unmodified mRNA (Left Panel) Contains natural uridine (U) as one of its nucleotide bases. When introduced into a cell, the immune system recognizes it as foreign, triggering an inflammatory response. The mRNA is translated into protein, but this immune activation can cause unwanted side effects. Base-Modified mRNA (Right Panel) Uridine (U) is replaced with pseudouridine (Ψ), a modified nucleoside. This modification reduces immune recognition, preventing the inflammatory response. Allows for more efficient translation and increased stability of the mRNA. Used in mRNA vaccines (e.g., COVID-19 vaccines by Pfizer & Moderna) to minimize immune-related side effects while enhancing protein expression. In further studies published in 2008 and 2010, Karikó and Weissman showed that the delivery of mRNA generated with base modifications markedly increased protein production compared to unmodified mRNA. The effect was due to the reduced activation of an enzyme that regulates protein production. Through their discoveries that base modifications both reduced inflammatory responses and increased protein production, Karikó and Weissman had eliminated critical obstacles on the way to clinical applications of mRNA.
  • #12 Mechanism of immune activation by mRNA-LNPs vaccines: Upon administration, mRNALNPs are internalized by immune cells, where the mRNA is released from the LNPs. The mRNA is then recognized by ribosomes, leading to the translation of antigenic proteins. These proteins are processed and degraded by proteasomes producing small peptides that then get loaded on major histocompatibility complex class I (MHC I) molecules and presented on the cell surface, which binds to the T cell receptor (TCR), activating CD8+ T cells to kill infected cells. The produced antigen can also be endocytosed by antigen-presenting cells (APCs) and further degraded by the lysosomes producing small peptides that then get loaded on major histocompatibility complex class II (MHC II) molecules for recognition by CD4+ T cells, which subsequently activate both the cellular immune responses by secreting inflammatory cytokines and the humoral immune responses by activating B cells.
  • #14 require longer production times. Live vaccines may not be suitable for immunocompromised individuals. Some vaccines require adjuvants to boost immune response. Advantages of Conventional Vaccines Over mRNA Vaccines Better Stability and Easier Storage Longer Shelf Life More Established Long-Term Safety Data No Need for Advanced Cold Chain Logistics Fewer Short-Term Side Effects Proven Effectiveness Across a Wide Range of Diseases No Risk of Rapid Degradation Well-Established Manufacturing & Production Facilities Longer-Lasting Immunity Less Risk of Overstimulation of the Immune System
  • #15 faster development, stronger immune responses, better safety, and easier large-scale production.
  • #21 mRNA vaccines require efficient delivery systems to protect the fragile mRNA from degradation and ensure successful entry into the target cells. polymer-based, peptide-based, and exosome-based systems
  • #22 Polymeric nanoparticles are used as alternative mRNA delivery systems to protect mRNA from degradation and enhance cellular uptake. Early systems using polyethylenimine (PEI) had toxicity issues, which were improved by PEGylation and cyclodextrin conjugation. Biodegradable polymers like poly(amino esters) and pH-responsive polymers enable controlled release, while amphipathic and dendrimer-based systems enhance mRNA binding and transfection. Modifications in polymer length, charge density, and composition further optimize efficacy, stability, and safety for mRNA vaccine applications. Polymer-Based Delivery Systems: offering better stability Polyethyleneimine (PEI) – Effective but can be toxic. Poly(β-amino esters) (PBAEs) – More biodegradable and less toxic than PEI. Chitosan Nanoparticles – Biocompatible, low toxicity, and enhances cellular uptake. ✔ More stable than lipid-based carriers, tunable for targeted delivery. ❌ Lower transfection efficiency, potential cytotoxicity.
  • #23 Peptide-Based Delivery Systems ✔ Uses cell-penetrating peptides (CPPs) to enhance mRNA uptake. CPPs interact electrostatically with mRNA to form nano-complexes for efficient delivery. Helps with endosomal escape, allowing mRNA release into the cytoplasm. ✔ Low toxicity and biocompatibility. ❌ Less efficient than lipid-based systems, requires chemical modification. 1️⃣ mRNA Encapsulation & Protection – Cationic peptides (e.g., arginine-rich peptides) electrostatically bind to negatively charged mRNA, forming stable nano-complexes that protect mRNA from degradation. 2️⃣ Cellular Uptake (Endocytosis & Membrane Fusion) – Peptide-mRNA complexes enter cells via endocytosis, and pH-sensitive cell-penetrating peptides (CPPs) like RALA peptides alter their structure in acidic endosomes, facilitating membrane fusion. 3️⃣ Endosomal Escape & mRNA Release – Peptides disrupt endosomal membranes, allowing mRNA release into the cytoplasm, preventing degradation and enabling translation.
  • #24 protamine-based mRNA delivery can induce strong anti-tumor immune responses and has been used in cancer therapy (glioblastoma) and vaccine development (rabies, influenza A).
  • #25 Lipid Nanoparticles (LNPs) are nanoscale lipid-based carriers that efficiently transport mRNA into the cytosol while protecting it from RNase degradation. LNPs facilitate intracellular delivery by fusing with the endosomal membrane, ensuring mRNA release into the cytoplasm for translation. They have been widely used in SARS-CoV-2, influenza, and tuberculosis mRNA vaccines due to their biocompatibility, modularity, and high payload capacity. LNPs typically consist of: ✔ Ionizable lipids (40–50%) – Aid in mRNA binding and endosomal escape. ✔ Cholesterol (38–45%) – Enhances membrane stability. ✔ Helper phospholipids (10–12%) – Assist in lipid bilayer fusion. ✔ PEGylated lipids (1–2%) – Improve circulation time and prevent aggregation. Cationic lipids like DOTMA, DOTAP, and Lipofectamine were first used for RNA delivery, later evolving into ionizable lipids (e.g., DODAP, DODMA, DLin-MC3-DMA) for higher efficacy and safety. Microfluidics technology is now revolutionizing LNP formulation by enabling precise mixing, homogeneous nanoparticle formation, and improved mRNA encapsulation. The final LNP-mRNA vaccine product undergoes filtration, buffer exchange (TFF), and sterilization, ensuring stability in either lyophilized or liquid format. LNPs remain the gold standard for mRNA vaccine delivery, with ongoing advancements improving stability, efficiency, and large-scale manufacturing.
  • #28 Organ-on-a-Chip (OoC) Technology Organ-on-a-chip (OoC) is an advanced microfluidic technology that mimics the structure and function of human organs outside the body. It consists of tiny, 3D tissue cultures grown on a chip-like device with microchannels, enabling researchers to study human-like responses to drugs, toxins, and diseases without using animals. How Does It Work? These chips are bioengineered using human cells from specific organs (e.g., lungs, liver, heart). Microfluidic channels simulate blood flow and mechanical forces similar to real organs. Sensors can monitor biological responses like oxygen levels, metabolism, and inflammation. Examples of Organ-on-a-Chip Models 🔬 Lung-on-a-Chip – Mimics breathing mechanics to study respiratory diseases and drug effects. 🧠 Brain-on-a-Chip – Models the blood-brain barrier for neurodegenerative disease research. 🫀 Heart-on-a-Chip – Simulates heart contractions and is used for cardiac drug testing. 🦠 Gut-on-a-Chip – Replicates the intestinal environment, useful for studying the microbiome and drug absorption. 🫁 Liver-on-a-Chip – Tests drug metabolism and toxicity before human trials. Advantages Over Traditional Models ✅ More accurate than animal models – Mimics human physiology better. ✅ Reduces animal testing – Ethical and cost-effective alternative. ✅ Faster drug development – Speeds up preclinical research. ✅ Personalized medicine potential – Uses patient-derived cells for tailored treatments. Future of Organ-on-a-Chip Multi-organ chips (Body-on-a-Chip) to simulate entire human systems. AI-driven analysis for real-time predictions. Integration into clinical trials for personalized drug screening.
  • #29 What are Exosomes? Exosomes are small extracellular vesicles (EVs) (30–150 nm) secreted by cells that play a crucial role in cell-to-cell communication by transporting proteins, lipids, RNA, and mRNA between cells. They originate from endosomal compartments and are released when multivesicular bodies (MVBs) fuse with the plasma membrane. Key Features of Exosomes: ✔ Naturally occurring – Derived from cells, making them highly biocompatible. ✔ Efficient cargo carriers – Transport mRNA, miRNA, proteins, and lipids for gene regulation. ✔ Low immunogenicity & toxicity – Suitable for drug delivery and vaccine applications. ✔ Targeted delivery – Surface ligands allow exosomes to interact with specific recipient cells. Applications of Exosomes in Medicine: mRNA vaccine delivery – An alternative to lipid nanoparticles (LNPs). Cancer therapy – Engineered exosomes for targeted drug delivery. Gene therapy – Delivering therapeutic RNA and CRISPR components. Diagnostics – Used as biomarkers for diseases like cancer and neurodegenerative disorders. Exosomes are being explored as next-generation drug and mRNA delivery vehicles, offering advantages over synthetic carriers due to their natural origin, biocompatibility, and targeting capabilities. 🚀
  • #32 mRNA technology is currently being explored for autoimmune and rare disease therapies, focusing on immune modulation, protein replacement, and enzyme restoration. In the future, personalized treatments, gene editing, tolerogenic vaccines, and improved delivery systems will expand its role in autoimmune conditions (RA, lupus, MS) and rare genetic disorders (DMD, CF, metabolic diseases). mRNA therapy for autoimmune diseases works by reprogramming the immune system to restore immune tolerance and prevent excessive immune responses. This is achieved by introducing specific mRNA sequences that encode tolerogenic proteins or anti-inflammatory factors to regulate the immune response.
  • #47 Before Phase 1, there are crucial preclinical phases that determine whether a drug is safe enough to be tested in humans. These include: 1. Discovery & Development Researchers identify potential drug candidates by studying disease mechanisms. New molecules are synthesized or discovered through screening. Drug formulations are developed for testing. 2. Preclinical Testing (In Vitro & In Vivo Studies) In vitro (test tube/cell culture studies): Drug effects are tested on isolated cells or tissues. In vivo (animal studies): Drug is tested in animals (typically mice, rats, or primates) to evaluate: Toxicity (safety profile) Pharmacokinetics (ADME: Absorption, Distribution, Metabolism, Excretion) Pharmacodynamics (mechanism of action & biological effects) Regulatory agencies (like the FDA, EMA, or CDSCO) require Good Laboratory Practice (GLP)-compliant studies. 3. Investigational New Drug (IND) Application If preclinical results are promising, researchers submit an IND application to regulatory authorities. The IND includes data on: Preclinical findings Proposed human trial design Manufacturing details Approval of the IND allows Phase 1 clinical trials to begin