View Article

Abstract

The P2X7 receptor (P2X7R) is a distinctive member of the purinergic receptor family, functioning as a ligand-gated ion channel that is predominantly activated by high concentrations of extracellular adenosine triphosphate (ATP). Unlike other P2X receptor subtypes, P2X7R exhibits unique biophysical and physiological properties, including the ability to form large non-selective membrane pores, initiate membrane blebbing, and trigger the release of pro-inflammatory cytokines such as interleukin-1? (IL-1?) and interleukin-18 (IL-18). These actions position P2X7R as a crucial mediator of immune cell activation, inflammation, and programmed cell death, particularly pyroptosis. Recent studies have underscored the receptor’s pivotal role in the pathogenesis of a wide array of diseases, including chronic inflammatory conditions (e.g., rheumatoid arthritis, Crohn’s disease), various cancers (e.g., glioblastoma, leukemia, breast cancer), and neurodegenerative disorders (e.g., Alzheimer’s disease, multiple sclerosis). The receptor's expression is often upregulated in disease states, correlating with disease severity and poor prognosis, making it a promising therapeutic target. This review presents a comprehensive analysis of the structural biology of P2X7R, its activation mechanisms, and downstream signaling cascades. We examine how P2X7R contributes to inflammation, immune surveillance, tumor progression, and neuronal injury. Furthermore, we detail the pharmacological agents developed to modulate P2X7R activity, including antagonists, monoclonal antibodies, and allosteric modulators. Several of these compounds have shown promising results in preclinical models and are currently undergoing clinical trials. Despite these advancements, targeting P2X7R therapeutically poses several challenges, such as receptor isoform variability, tissue-specific expression, and the dual role of P2X7R in promoting both cell survival and death. We discuss these challenges in depth and highlight the opportunities for future research, including biomarker-guided therapies and combination approaches. In summary, the P2X7 receptor represents a novel and versatile pharmacological target with immense potential for treating a broad spectrum of inflammatory, oncological, and neurological diseases. Continued investigation into the receptor’s biology and the refinement of its modulators could unlock transformative therapeutic strategies in modern medicine.

Keywords

P2X7 receptor (P2X7R), Purinergic signaling, Inflammation, Neuroinflammation, Cancer immunology, Apoptosis, Pyroptosis, NLRP3 inflammasome, ATP-gated ion channel

Introduction

Purinergic signaling, a form of extracellular nucleotide-mediated communication, plays a pivotal role in maintaining physiological homeostasis and orchestrating responses to pathological stimuli. Adenosine triphosphate (ATP), traditionally recognized as an intracellular energy currency, also acts as a critical extracellular signaling molecule upon release from damaged, stressed, or activated cells. Once in the extracellular space, ATP binds to and activates a family of purinergic receptors, broadly categorized into P1 (adenosine receptors) and P2 receptors, the latter subdivided into P2X ionotropic and P2Y metabotropic subtypes. Among the P2X receptor family, the P2X7 receptor (P2X7R) stands out due to its unique structural and functional characteristics. Unlike other P2X receptors that mediate rapid ion fluxes in response to low ATP concentrations, P2X7R requires significantly higher ATP levels—typically in the hundreds of micromolar range—for activation. Such elevated extracellular ATP concentrations are commonly observed in pathological contexts including infection, inflammation, ischemia, trauma, and within the tumor microenvironment. P2X7R is predominantly expressed on cells of the immune system, including macrophages, monocytes, dendritic cells, and microglia, where it contributes to the modulation of innate and adaptive immune responses. Activation of this receptor leads to the opening of a cation-selective channel and, upon prolonged stimulation, the formation of a large transmembrane pore permeable to molecules up to 900 Da. This phenomenon is associated with the induction of various downstream processes such as NLRP3 inflammasome activation, caspase-1 cleavage, interleukin-1β (IL-1β) secretion, reactive oxygen species (ROS) generation, and eventually, cell death via pyroptosis or apoptosis.

 

 

 

 

Figure 1. Prospective applications of the P2X7R in clinical diagnosis

 

Given its central role in regulating immune responses and inflammation, P2X7R has gained considerable attention as a drug target for a broad spectrum of diseases. In inflammatory disorders, P2X7R activation exacerbates tissue damage and chronic immune activation. Conversely, in oncology, the receptor plays a paradoxical role—facilitating tumor growth in some contexts by promoting immune evasion and angiogenesis, while in others, enhancing anti-tumor immunity through immunogenic cell death. Similarly, in neurological diseases, microglial P2X7R contributes to neuroinflammation and neuronal damage, offering opportunities for neuroprotective interventions. This review aims to provide a detailed overview of the structure, activation, and downstream signaling mechanisms of P2X7R, its role in inflammation, cancer, and neurological disorders, and the current status of pharmacological efforts to modulate its activity. By understanding the multifaceted roles of P2X7R in health and disease, we can better harness its potential as a therapeutic target in modern pharmacology.

2. Structure and Activation Mechanism of P2X7 Receptors

The P2X7 receptor (P2X7R) is a unique member of the P2X family of purinergic receptors, which are ligand-gated ion channels activated by extracellular adenosine triphosphate (ATP). Unlike other P2X receptors, P2X7R exhibits distinct structural and functional characteristics that endow it with a dual gating mechanism and a pivotal role in immune signaling and cell death pathways.

Structural Composition

P2X7R is a trimeric protein, with each subunit comprising two transmembrane domains (TM1 and TM2), a large extracellular loop containing the ATP-binding site, and both N- and C-terminal intracellular domains. Upon trimerization, the receptor forms a central ion-conducting pore. One of the hallmark structural features of P2X7R is its exceptionally long C-terminal intracellular tail, which is not present in other P2X family members. This region is critical for downstream signaling interactions, particularly those involving immune and inflammatory responses.

Activation by ATP

Under physiological conditions, low extracellular ATP concentrations can activate P2X7R to function as a non-selective cation channel, allowing the influx of Na? and Ca²?, and the efflux of K?. This ionic flux contributes to membrane depolarization and initiates various cellular responses, including activation of downstream kinases and transcription factors. However, when P2X7R is exposed to sustained or high concentrations of ATP, typically observed in pathological conditions such as infection, inflammation, or tissue injury, the receptor undergoes a conformational change that converts it into a macropore. This large, dilated pore is permeable to hydrophilic molecules with molecular weights up to approximately 900 Da, including dyes like YO-PRO-1 and ethidium bromide.

Macropore Formation and Cellular Consequences

The transition from ion channel to macropore is a defining feature of P2X7R and is closely associated with cellular stress and danger signaling. Macropore formation leads to:

  • Cell swelling due to osmotic imbalance
  • Plasma membrane rupture, allowing the release of intracellular contents
  • Activation of apoptotic or pyroptotic pathways depending on the cellular context

This function places P2X7R at the crossroads of regulated cell death and immune activation.

Interaction with the Inflammasome

P2X7R has been shown to directly interact with components of the NLRP3 inflammasome, particularly in macrophages and microglia. The K? efflux caused by P2X7R activation is a well-recognized trigger for NLRP3 inflammasome assembly, leading to the activation of caspase-1 and the maturation and secretion of pro-inflammatory cytokines IL-1β and IL-18. Thus, P2X7R serves as a critical link between extracellular ATP signals and innate immune responses.

Key Features of P2X7R Activation:

  • Long intracellular C-terminal tail: Essential for protein-protein interactions and downstream signaling, particularly with immune-related pathways.
  • Ion channel to macropore transition: Occurs upon prolonged or high-dose ATP stimulation, enabling the passage of large molecules.
  • Inflammasome activation: Facilitates assembly of the NLRP3 inflammasome complex through K? efflux and protein interaction.
  • Regulation of cell death: Promotes pyroptosis or apoptosis depending on context and co-signals.

3. Role in Inflammation and Immune Response

The P2X7 receptor (P2X7R) plays a pivotal role in orchestrating inflammatory and immune responses. Its expression is primarily localized in immune cells such as macrophages, dendritic cells, and microglia, where it acts as a key sensor of extracellular ATP—a "danger signal" released during cellular stress, injury, or infection.

Mechanistic Role in Inflammation

Upon activation by high levels of extracellular ATP, P2X7R initiates a cascade of intracellular events that shape both innate and adaptive immunity:

  • NLRP3 Inflammasome Activation: One of the hallmark roles of P2X7R is its involvement in the activation of the NLRP3 inflammasome. This is triggered by potassium efflux through P2X7R, leading to the recruitment of adaptor proteins and pro-caspase-1, which subsequently becomes activated. Active caspase-1 cleaves pro-IL-1β and pro-IL-18 into their mature, bioactive forms, facilitating their secretion and amplifying the inflammatory response.
  • Reactive Oxygen Species (ROS) Production: P2X7R stimulation has been linked to mitochondrial dysfunction and oxidative stress, resulting in the generation of ROS. These species further enhance inflammatory signaling and can damage cellular components, contributing to tissue pathology.
  • Induction of Cell Death: Under conditions of sustained activation, P2X7R forms large pores that compromise membrane integrity, leading to apoptosis, pyroptosis, or necrosis. This promotes the release of damage-associated molecular patterns (DAMPs), which further propagate inflammation, especially in chronic disease states.

Clinical Relevance in Inflammatory Disorders

Due to its strong link to immune activation and pro-inflammatory effects, P2X7R is increasingly recognized as a therapeutic target in multiple inflammatory and autoimmune diseases:

  • Rheumatoid Arthritis (RA): In the inflamed synovial tissue of RA patients, elevated extracellular ATP levels are observed. Overactivation of P2X7R contributes to the recruitment and activation of immune cells, leading to synovial inflammation, joint degradation, and pain. Antagonists of P2X7R have shown promise in experimental arthritis models.
  • Inflammatory Bowel Disease (IBD): In Crohn’s disease and ulcerative colitis, P2X7R activation exacerbates gut mucosal inflammation through cytokine release and epithelial barrier disruption. It also promotes macrophage and dendritic cell infiltration, worsening mucosal damage.
  • Sepsis and Infectious Diseases: During infections, P2X7R helps mount an effective immune response by stimulating phagocyte activation and cytokine secretion. However, overactivation in sepsis may lead to uncontrolled systemic inflammation, organ dysfunction, and poor outcomes, highlighting the need for balanced modulation.

Summary of Key Functions:

  • Pro-inflammatory cytokine release: IL-1β, IL-18 via NLRP3 inflammasome
  • Oxidative stress induction: ROS generation through mitochondrial pathways
  • Immunogenic cell death: Via macropore formation and DAMPs release

4. P2X7 Receptors in Cancer

The involvement of P2X7 receptors (P2X7R) in cancer is a subject of increasing scientific interest, marked by dual and context-dependent roles in tumor biology. Depending on the tumor type, microenvironment, and degree of receptor activation, P2X7R can exhibit both tumor-suppressive and tumor-promoting functions.

Tumor-Suppressive Functions of P2X7R

Under certain conditions, particularly when receptor activation is acute or high in intensity, P2X7R contributes to anti-tumor mechanisms through:

  • Induction of Cell Death: P2X7R-mediated macropore formation can lead to apoptosis or pyroptosis in tumor cells, especially when high extracellular ATP levels overwhelm tumor defenses. This contributes to the elimination of malignant cells.
  • Immune Activation: By triggering IL-1β and IL-18 release through NLRP3 inflammasome activation, P2X7R can enhance anti-tumor immunity, promoting immune cell recruitment and activation against tumor-associated antigens.
  • Immunogenic Cell Death (ICD): The release of damage-associated molecular patterns (DAMPs) such as ATP and HMGB1 following P2X7R activation may promote dendritic cell maturation and T-cell activation, contributing to adaptive anti-tumor responses.

Tumor-Promoting Roles of P2X7R

Conversely, in many malignancies, chronic or sustained activation of P2X7R can facilitate tumor progression through several mechanisms:

  • Cell Proliferation and Survival: Tumor cells can exploit low-level P2X7R signaling to support mitogenic pathways, contributing to proliferation, metabolic reprogramming, and resistance to cell death.
  • Angiogenesis and Invasion: P2X7R activation has been shown to upregulate vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs), aiding angiogenesis and extracellular matrix degradation, respectively—key steps in tumor invasion and metastasis.
  • Immune Evasion: Upregulation of P2X7R in tumor-infiltrating immune cells can lead to a paradoxical immunosuppressive microenvironment, through chronic inflammation, T-cell exhaustion, or regulatory cell recruitment, allowing tumor cells to evade immune destruction.

Representative Cancer Types and Evidence

  • Breast Cancer: Studies have shown that P2X7R is overexpressed in more aggressive breast cancer subtypes. Its activation correlates with enhanced migration, invasiveness, and poor prognosis. Conversely, full activation leading to pore formation can reduce viability of these cells, emphasizing its duality.
  • Glioblastoma and Melanoma: In these highly aggressive cancers, autocrine/paracrine ATP signaling sustains P2X7R activity, promoting tumor progression. In glioblastoma, P2X7R supports the maintenance of the cancer stem-like cell niche and contributes to resistance to chemotherapy.

Summary of Context-Dependent Roles

 

Function

Tumor-Suppressive Role

Tumor-Promoting Role

Cell Death

Induction of apoptosis/pyroptosis

Resistance to death, survival advantage

Immune Modulation

Pro-inflammatory cytokines, immune activation

Immune suppression, evasion

Microenvironment

Release of DAMPs for ICD

Promotion of angiogenesis, invasion

ATP Levels

High ATP causes cell death

Chronic ATP supports growth

 

The therapeutic targeting of P2X7R in cancer remains a promising yet challenging approach. Future strategies may focus on modulating P2X7R activation thresholds or developing selective agonists/antagonists to exploit its tumor-suppressive properties while minimizing its oncogenic potential.

5. Neurological Implications

The P2X7 receptor (P2X7R) plays a critical role in the central nervous system (CNS), particularly in mediating neuroinflammation and glial cell function. It is predominantly expressed in microglia, the resident immune cells of the brain, and to a lesser extent in astrocytes and oligodendrocytes. Its activation by elevated extracellular ATP—a common feature of neurological injury or disease—leads to profound effects on neuronal health, glial activity, and synaptic regulation.

Role in Neuroinflammation and Glial Activation

P2X7R activation in the CNS initiates a robust inflammatory response, characterized by the release of pro-inflammatory cytokines such as IL-1β, IL-18, and TNF-α. This occurs primarily through NLRP3 inflammasome activation in microglia, resulting in chronic inflammation, glial scarring, and neuronal damage. Furthermore, sustained activation promotes ROS generation, blood-brain barrier (BBB) disruption, and synaptic dysfunction, exacerbating the progression of neurodegenerative diseases.

Key Neurological Disorders Involving P2X7R

  • Alzheimer’s Disease (AD): In AD, P2X7R is upregulated in activated microglia surrounding amyloid-beta (Aβ) plaques. Its activation amplifies Aβ-induced inflammation, leading to increased cytokine production and further neuronal loss. Additionally, P2X7R-mediated ATP signaling has been implicated in synaptic dysfunction and cognitive impairment in AD models.
  • Parkinson’s Disease (PD): Elevated P2X7R expression has been observed in both microglia and astrocytes in PD-affected brain regions. Its activation contributes to the degeneration of dopaminergic neurons in the substantia nigra, likely through inflammatory and oxidative pathways. P2X7R inhibitors have shown neuroprotective effects in experimental models.
  • Multiple Sclerosis (MS): P2X7R is involved in demyelination and neurodegeneration seen in MS. It is upregulated in active MS lesions and contributes to T cell infiltration, microglial activation, and myelin sheath destruction. Its role in BBB disruption and inflammation makes it a potential target for therapeutic intervention in MS.
  • Neuropathic Pain: P2X7R plays a key role in glial activation in the spinal cord, which is a major contributor to chronic pain. Upon nerve injury, ATP release activates P2X7R in spinal microglia and astrocytes, leading to the release of pro-nociceptive cytokines that sensitize pain pathways and enhance pain perception. Antagonism of P2X7R has shown promise in alleviating neuropathic pain in preclinical studies.

Summary of P2X7R in CNS Disorders

Disorder

P2X7R-Related Effects

Alzheimer’s Disease

Enhances Aβ-induced microglial activation and cytokine release

Parkinson’s Disease

Promotes dopaminergic neuron death via neuroinflammation

Multiple Sclerosis

Drives demyelination, T cell activation, and BBB breakdown

Neuropathic Pain

Triggers spinal glial activation and cytokine-mediated sensitization

Targeting P2X7R in the CNS presents a promising therapeutic avenue. However, due to its complex role in immune defense and cell viability, strategies must be precisely tailored to disease stage and severity to avoid adverse effects on normal neuroimmune functions.

6. Pharmacological Modulators of P2X7 Receptors

Given the involvement of P2X7 receptors (P2X7R) in a wide range of pathological conditions—including chronic inflammation, neurodegeneration, autoimmune disorders, and cancer—numerous pharmacological strategies have been pursued to modulate its activity. These strategies include small-molecule antagonists, allosteric modulators, biologics, and gene-silencing approaches.

6.1 Antagonists

The most extensively studied class of P2X7 modulators are antagonists, which inhibit receptor activation by extracellular ATP. These agents are under investigation for various autoimmune, inflammatory, and neurological diseases.

 

Drug Candidate

Class

Notes/Applications

AZD9056

Competitive antagonist

Developed by AstraZeneca; evaluated in rheumatoid arthritis (RA) and Crohn’s disease; demonstrated safety but limited efficacy in late-phase trials.

GSK1482160

Brain-penetrant antagonist

Developed by GlaxoSmithKline; crosses the blood-brain barrier and was evaluated for neuropathic pain and multiple sclerosis in preclinical studies.

JNJ-47965567

Potent and selective

A highly selective P2X7R antagonist with oral bioavailability; showed efficacy in rodent pain models and potential use in psychiatric disorders.

A-438079

Non-brain penetrant

Widely used in experimental models of peripheral inflammation and immune activation; does not cross the BBB, limiting its CNS application.

 

 

These antagonists work primarily by blocking ATP binding or preventing conformational changes necessary for pore formation and ion channel function.

6.2 Positive Allosteric Modulators (PAMs)

Positive allosteric modulators enhance the receptor’s sensitivity to ATP, increasing its activation under specific physiological or pathological conditions. While less explored than antagonists, PAMs are gaining interest in fields like immuno-oncology, where enhanced P2X7R activation in tumor-infiltrating immune cells could boost anti-tumor immunity.

  • Mechanism: PAMs bind to allosteric sites distinct from the ATP-binding pocket, altering receptor conformation to increase ATP efficacy.
  • Challenges: Balancing activation without inducing cytotoxic pore formation remains a hurdle in clinical translation.

6.3 Biologics and RNA Interference Approaches

In addition to small molecules, biological agents and gene-silencing technologies have emerged as promising tools for selectively targeting P2X7R expression or function:

  • Monoclonal Antibodies: Antibody-based inhibition of P2X7R offers specificity but is currently limited by poor tissue and CNS penetration. Most are still in preclinical development.
  • siRNA and Antisense Oligonucleotides (ASOs): These nucleic acid-based therapies are designed to downregulate P2X7R mRNA, effectively reducing receptor expression. These approaches have shown promise in animal models of neuroinflammation and autoimmune disorders, with ongoing research on improving delivery systems and stability.

Summary of Pharmacological Modulation Strategies

 

Modulator Type

Examples

Applications

Small-molecule antagonists

AZD9056, GSK1482160, JNJ-47965567

Autoimmune diseases, pain, CNS disorders

Positive allosteric modulators

Under investigation

Cancer immunotherapy, immune modulation

Biologics

Anti-P2X7R monoclonal antibodies

High specificity; limited by poor penetration

RNA interference

siRNA, ASOs

Gene-level suppression of P2X7R; experimental stages

 

P2X7R remains a highly drugable target, but its dual physiological roles in immune surveillance and cell death necessitate careful therapeutic modulation to avoid unintended side effects. The future lies in precision-targeted approaches, potentially using nanocarriers, tissue-specific delivery, or context-dependent activation/inhibition strategies.

7. Clinical Trials and Therapeutic Potential

The therapeutic targeting of P2X7 receptors (P2X7R) has garnered considerable interest due to their involvement in a wide range of inflammatory, autoimmune, neurological, and oncological conditions. Despite strong preclinical efficacy, the clinical translation of P2X7R antagonists has faced significant hurdles, with few candidates advancing to late-phase clinical trials.

Barriers to Clinical Success

Several challenges have limited the progression of P2X7R-targeted therapies:

  • Species-Specific Differences: The structure and pharmacology of P2X7R vary significantly between rodents (used in preclinical models) and humans. These differences can affect drug binding affinity, efficacy, and downstream signaling, resulting in poor translational outcomes.
  • Compensatory Pathways: The immune system possesses redundant signaling networks, and inhibition of P2X7R may lead to compensatory activation of other purinergic or inflammatory receptors, diminishing the therapeutic effect.
  • Dual and Context-Dependent Roles: P2X7R can exert both pro-inflammatory and immunosuppressive effects depending on the cell type and disease stage. In cancer, for instance, P2X7R may either enhance immune surveillance or promote tumor progression. These ambiguous roles complicate therapeutic targeting and risk-benefit evaluation.

Ongoing Areas of Clinical Investigation

Despite these limitations, research into P2X7R modulation continues across several therapeutic areas:

  • Autoimmune Disorders: Conditions like rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) were among the first indications for P2X7R antagonists. AZD9056 and other small molecules have undergone Phase I/II trials, showing safety but limited efficacy, possibly due to the issues outlined above.
  • Chronic Pain: Preclinical models have demonstrated that P2X7R antagonists alleviate neuropathic and inflammatory pain by inhibiting microglial activation in the spinal cord. Brain-penetrant compounds like GSK1482160 and JNJ-47965567 are under evaluation for pain disorders and migraine.
  • Neurodegenerative Conditions: In Alzheimer’s, Parkinson’s, and multiple sclerosis, P2X7R contributes to microglial activation, neuroinflammation, and neuronal loss. Ongoing research is exploring the use of P2X7R inhibitors to preserve cognitive function and delay disease progression.
  • Adjunctive Cancer Therapies: While not yet a primary target in oncology, P2X7R modulation is being studied as an adjunctive approach to enhance anti-tumor immunity or reduce cancer-related inflammation. Modulation strategies may include inhibitors in tumor-promoting settings or positive allosteric modulators in immuno-oncology contexts.

Summary: Therapeutic Outlook

 

Domain

Clinical Focus

Status/Challenges

Autoimmune diseases

RA, IBD

Early trials; mixed efficacy

Chronic pain

Neuropathic, inflammatory pain

Promising preclinical results; brain-penetration needed

Neurodegeneration

AD, PD, MS

Investigational; requires long-term safety data

Oncology

Adjunct to immunotherapy

Under exploration; dual role complicates application

 

The therapeutic potential of P2X7R modulators remains significant but requires refined targeting strategies, biomarker-guided patient selection, and possibly combination therapies to overcome biological complexity and enhance clinical efficacy.

8. Challenges and Future Directions

While the P2X7 receptor (P2X7R) remains a highly promising therapeutic target due to its involvement in inflammation, immunity, neurodegeneration, and cancer, several critical challenges continue to impede the full clinical translation of P2X7R-targeted therapies. Addressing these limitations is essential for realizing the receptor’s therapeutic potential across multiple disease domains.

Key Challenges in P2X7R Drug Development

  • Selectivity and Specificity

One of the foremost obstacles is achieving high selectivity for P2X7R without cross-reactivity with other P2X family members (e.g., P2X1–P2X6), which could lead to off-target effects. The close structural homology among P2X receptors makes it difficult to develop antagonists or modulators that are both potent and receptor-specific, especially across species.

  • Blood-Brain Barrier (BBB) Penetration
    Many small-molecule antagonists demonstrate limited CNS penetration, which restricts their use in treating neurological and neuroinflammatory disorders such as Alzheimer’s, Parkinson’s, and multiple sclerosis. The development of brain-penetrant compounds remains a priority for CNS-related applications.
  • Risk of Immunosuppression

P2X7R is a key regulator of host defense mechanisms. Chronic or systemic inhibition may impair innate immune responses, increasing the risk of infections or compromising anti-tumor immunity. Balancing therapeutic efficacy with immune competence is a critical consideration, especially in long-term treatments.

  • Lack of Predictive Biomarkers

The heterogeneous expression and activation of P2X7R across tissues and disease states necessitate the development of biomarkers that can identify patients with upregulated P2X7R signaling. Such biomarkers would enable precision medicine approaches, ensuring that only those likely to benefit receive P2X7R-targeted therapies.

Future Directions

To overcome these challenges and fully harness the therapeutic promise of P2X7R, future research should focus on the following strategies:

  1. Structure-Based Drug Design
    Advances in cryo-electron microscopy (cryo-EM) and computational modeling are enabling a better understanding of P2X7R structure and conformational states, facilitating the design of more selective and effective modulators.
  2. Development of CNS-Active Compounds
    The synthesis of lipophilic, low-molecular-weight antagonists capable of crossing the BBB will be crucial for expanding the use of P2X7R inhibitors in neurodegenerative and psychiatric conditions.
  3. Context-Dependent Modulation
    Instead of full inhibition, partial antagonists or biased modulators may allow for disease-specific regulation of P2X7R without compromising its physiological functions.
  4. Combination Therapies
    Combining P2X7R modulators with anti-inflammatory drugs, immune checkpoint inhibitors, or chemotherapy could enhance efficacy and reduce the need for high-dose monotherapies.
  5. Identification of Biomarkers and Diagnostic Tools
    Integration of transcriptomic, proteomic, and imaging techniques could aid in identifying reliable biomarkers to track P2X7R expression/activity and predict therapeutic response.

Summary of Key Needs and Innovations

Challenge

Proposed Solution

Selectivity issues

High-resolution structural studies and rational drug design

Limited CNS penetration

Develop BBB-permeable antagonists

Risk of immunosuppression

Use context-specific or partial modulators

Absence of biomarkers

Identify molecular or imaging-based diagnostic indicators

In conclusion, while significant obstacles remain, ongoing advancements in molecular pharmacology, drug delivery, and personalized medicine are likely to transform P2X7R from a promising target into a clinically validated therapeutic axis across multiple disease areas.

9. CONCLUSION

The P2X7 receptor (P2X7R) stands out as a unique and multifunctional pharmacological target, playing pivotal roles in inflammatory, oncological, and neurological disorders. Its capacity to function both as a non-selective ion channel and as a macropore-forming receptor allows it to participate in a wide spectrum of physiological and pathological processes—from immune cell activation and cytokine release to cell death and tissue remodeling. Importantly, P2X7R exhibits a dual role: it can be protective by enhancing host defense and promoting immunogenic cell death, yet also pathogenic when overactivated, contributing to chronic inflammation, tissue damage, or tumor progression. This duality underscores the need for precise and context-specific modulation of its activity in therapeutic settings. Despite promising preclinical findings, the translation of P2X7R-targeted therapies into clinical success has been limited by challenges such as species-specific receptor differences, blood-brain barrier penetration, and context-dependent immune effects. However, ongoing advances in structural biology, drug design, and biomarker discovery are steadily overcoming these barriers.

Future therapeutic strategies are likely to focus on:

  • Selective and brain-penetrant modulators with improved specificity
  • Combination therapies that integrate P2X7R modulation with standard treatments
  • Personalized approaches guided by receptor expression profiles and patient-specific biomarkers

Continued research into P2X7R signaling mechanisms, structural dynamics, and tissue-specific roles will be critical in harnessing its full therapeutic potential. As our understanding deepens, P2X7R is poised to become a cornerstone in the treatment of complex diseases at the intersection of immunity, inflammation, and degeneration.

REFERENCES

  1. Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S. The P2X7 Receptor in Infection and Inflammation. Immunity. 2017;47(1):15–31.
  2. Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: Recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 2014;66(3):638–75.
  3. Burnstock G. Introduction to purinergic signaling in the brain. Adv Exp Med Biol. 2013;986:1–12.
  4. Savio LEB, Coutinho-Silva R. Immunomodulatory effects of P2X7 receptor signaling during infectious diseases. Front Immunol. 2019;10:2238.
  5. Adinolfi E, Giuliani AL, De Marchi E, Pegoraro A, Orioli E, Di Virgilio F. The P2X7 receptor: A main player in inflammation. Biochem Pharmacol. 2018;151:234–44.
  6. Jiang LH. P2X7 receptor channel: Recent developments and perspectives. Purinergic Signal. 2015;11(4):411–8.
  7. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science. 1996;272(5262):735–8.
  8. Pelegrin P, Surprenant A. Pannexin-1 mediates large pore formation and interleukin-1β release by the ATP-gated P2X7 receptor. EMBO J. 2006;25(21):5071–82.
  9. He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–21.
  10. Wiley JS, Sluyter R, Gu BJ, Stokes L, Fuller SJ. The human P2X7 receptor and its role in innate immunity. Tissue Antigens. 2011;78(5):321–32.
  11. Barden JA, Bennett MR. Purinergic signaling and pain: The P2X3 and P2X7 receptor connection. Neuroscientist. 2000;6(3):209–14.
  12. Turner CM, Tam FW, Lai TS, Tarzi R, Burnstock G, Pusey CD, et al. Increased expression of the proapoptotic P2X7 receptor in glomerulonephritis. Kidney Int. 2004;66(2):610–20.
  13. Sluyter R, Stokes L. Significance of P2X7 receptor variants in human health and disease. Purinergic Signal. 2011;7(3):283–98.
  14. Monif M, Burnstock G, Williams DA. Microglia: Proliferation and activation driven by the P2X7 receptor. Int J Biochem Cell Biol. 2010;42(11):1753–6.
  15. Takenouchi T, Sekiyama A, Sekigawa A, Fujita M, Waragai M, Sugama S, et al. P2X7 receptor signaling pathway as a therapeutic target for neurodegenerative diseases. Arch Immunol Ther Exp. 2010;58(2):91–6.
  16. Skaper SD, Debetto P, Giusti P. The P2X7 purinergic receptor: From physiology to neurological disorders. FASEB J. 2010;24(2):337–45.
  17. Martin TA, Harrison G, Mansel RE, Jiang WG. The role of P2X7 in cancer: A brief review. J Oncol. 2009;2009:481–87.
  18. Amoroso F, Capece M, Rotondo A, Cangelosi D, Ferracin M, Franceschini A, et al. P2X7 receptor is a cytotoxic receptor selectively expressed on chronic lymphocytic leukemia B cells. Blood. 2015;125(21):346–55.
  19. Adinolfi E, Raffaghello L, Giuliani AL, Cavazzini F, Capece M, Chiozzi P, et al. Expression of P2X7 receptor increases in vivo tumor growth. Cancer Res. 2012;72(12):2957–69.
  20. Di Virgilio F, Vuerich M. Purinergic signaling in the immune system. Auton Neurosci. 2015;191:117–23.
  21. Rissiek B, Haag F, Boyer O, Koch-Nolte F. P2X7 on mouse T cells: One channel, many functions. Front Immunol. 2015;6:204.
  22. Francistiová L, Bianchi C, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer. 2023;23:85–104.
  23. Giuliani AL, Colognesi D, Ricco T, Roncato C, Capece M, Amoroso F, et al. Trophic activity of a naturally occurring truncated isoform of the P2X7 receptor. FASEB J. 2019;33(1):10285–98.
  24. Lees RM, Babon JJ, Brill AL, Hume DA, Stacey KJ, Hamilton JA. P2X7 receptor-deficient mice exhibit impaired osteoclast formation and altered inflammatory responses. J Immunol. 2014;193(2):871–80.
  25. Douguet L, Janho Dit Hreich S, Benzaquen J, Grutter T. Structural basis of P2X7 receptor activation and inhibition. Nat Commun. 2021;12(1):5183.
  26. Ulmann L, Levavasseur F, Avignone E, Peyroutou R, Hirbec H, Audinat E, et al. Involvement of P2X7 receptor in chronic pain models. J Neurosci. 2008;28(44):11106–18.
  27. Sperlágh B, Illes P. P2X7 receptor: An emerging target in CNS diseases. Trends Pharmacol Sci. 2014;35(10):537–47.
  28. Danquah W, Meyer-Schwesinger C, Rissiek B, Csoka B, Scholz K, Schnurr M, et al. Targeting the P2X7 receptor in inflammatory diseases. Expert Opin Ther Targets. 2016;20(4):453–67.
  29. Bhaskaracharya A, Dao-Ung P, Jalilian I, Spildrejorde M, Skarratt KK, Fuller SJ, et al. Pro-inflammatory role of the P2X7 receptor in an animal model of multiple sclerosis. J Neuroinflammation. 2014;11(1):137.
  30. Norenberg W, Gebicke-Haerter PJ, Illes P. Expression of P2X7 receptors on activated microglia in the rat spinal cord following peripheral nerve injury. Brain Res. 2001;926(1–2):206–7.
  31. North RA. Molecular physiology of P2X receptors. Physiol Rev. 2002;82(4):1013–67.
  32. Sluyter R. The P2X7 receptor. Adv Exp Med Biol. 2012;986:83–98.
  33. Gu BJ, Saunders BM, Jursik C, Wiley JS. The P2X7 receptor: A target for new anti-inflammatory therapies. Expert Opin Ther Targets. 2011;15(10):1137–50.
  34. Baroja-Mazo A, Martín-Sánchez F, Gomez AI, Martinez CM, Amores-Iniesta J, Compan V, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014;15(8):738–48.
  35. Keane RW, Dietrich WD, de Rivero Vaccari JP. Inflammasome proteins as biomarkers of multiple sclerosis. Front Neurol. 2018;9:135.
  36. Alves LA, Bezerra RJ, Faria RX, Ferreira LG, da Silva Frutuoso V. Physiological roles and potential therapeutic applications of the P2X7 receptor in inflammation and pain. Mem Inst Oswaldo Cruz. 2013;108(3):341–50.
  37. Nicke A. Homotrimeric complexes formed by the ATP-gated P2X1 receptor. J Biol Chem. 2003;278(13):10419–26.
  38. McGaraughty S, Wismer CT, Zhu CZ, Mikusa J, Honore P, Chu KL, et al. P2X7 receptor antagonism improves sciatic nerve-injury-induced neuropathic pain. Pain. 2007;131(1–2):229–43.
  39. Csóka B, Németh ZH, Szabó I, Davies DL, Vizi ES, Pacher P, et al. Adenosine A2A receptors control IL-10 production by macrophages and attenuate P2X7 receptor-stimulated IL-1β production. J Immunol. 2007;179(12):8280–6.
  40. Hafner M, Koeppl H, Misawa R, Duenas M, Jain A. Quantitative systems pharmacology analysis of ATP-mediated immune signaling in cancer immunotherapy. Cell Rep. 2022;39(9):110900.
  41. Ribeiro DE, Roncalho AL, Glaser T, Ulrich H, Lameu C. The P2X7 receptor in neurodegenerative diseases. Front Cell Neurosci. 2019;13:24.
  42. Illes P, Khan TM, Rubini P. Neuronal P2X7 receptors revisited: Do they really exist? J Neurosci. 2017;37(30):7049–62.
  43. Qu Y, Misaghi S, Newton K, Gilmour LL, Louie S, Cupp JE, et al. P2X7 receptor-stimulated cytokine secretion requires PI3K-dependent NF-κB activation. J Biol Chem. 2007;282(17):12507–18.
  44. Menzies RI, Unwin RJ, Bailey MA. Renal P2 receptors and hypertension. Acta Physiol (Oxf). 2015;213(1):232–41.
  45. Sluyter R, Fuller SJ, Burnstock G. P2X7 receptors in hematopoietic and immune system cells. Purinergic Signal. 2014;10(4):659–64.
  46. Amoroso F, Falzoni S, Adinolfi E, Ferrari D, Di Virgilio F. The P2X7 receptor is a key modulator of the PI3K/GSK-3β axis in microglia. J Neuroinflammation. 2020;17(1):203.
  47. Fang F, Pan J, Xu L, Yu Y, Lu Y, Sun B, et al. Inhibition of P2X7 receptor ameliorates sepsis-induced acute kidney injury in mice. Am J Physiol Renal Physiol. 2021;320(3):F409–21.

Reference

  1. Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S. The P2X7 Receptor in Infection and Inflammation. Immunity. 2017;47(1):15–31.
  2. Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: Recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 2014;66(3):638–75.
  3. Burnstock G. Introduction to purinergic signaling in the brain. Adv Exp Med Biol. 2013;986:1–12.
  4. Savio LEB, Coutinho-Silva R. Immunomodulatory effects of P2X7 receptor signaling during infectious diseases. Front Immunol. 2019;10:2238.
  5. Adinolfi E, Giuliani AL, De Marchi E, Pegoraro A, Orioli E, Di Virgilio F. The P2X7 receptor: A main player in inflammation. Biochem Pharmacol. 2018;151:234–44.
  6. Jiang LH. P2X7 receptor channel: Recent developments and perspectives. Purinergic Signal. 2015;11(4):411–8.
  7. Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science. 1996;272(5262):735–8.
  8. Pelegrin P, Surprenant A. Pannexin-1 mediates large pore formation and interleukin-1β release by the ATP-gated P2X7 receptor. EMBO J. 2006;25(21):5071–82.
  9. He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–21.
  10. Wiley JS, Sluyter R, Gu BJ, Stokes L, Fuller SJ. The human P2X7 receptor and its role in innate immunity. Tissue Antigens. 2011;78(5):321–32.
  11. Barden JA, Bennett MR. Purinergic signaling and pain: The P2X3 and P2X7 receptor connection. Neuroscientist. 2000;6(3):209–14.
  12. Turner CM, Tam FW, Lai TS, Tarzi R, Burnstock G, Pusey CD, et al. Increased expression of the proapoptotic P2X7 receptor in glomerulonephritis. Kidney Int. 2004;66(2):610–20.
  13. Sluyter R, Stokes L. Significance of P2X7 receptor variants in human health and disease. Purinergic Signal. 2011;7(3):283–98.
  14. Monif M, Burnstock G, Williams DA. Microglia: Proliferation and activation driven by the P2X7 receptor. Int J Biochem Cell Biol. 2010;42(11):1753–6.
  15. Takenouchi T, Sekiyama A, Sekigawa A, Fujita M, Waragai M, Sugama S, et al. P2X7 receptor signaling pathway as a therapeutic target for neurodegenerative diseases. Arch Immunol Ther Exp. 2010;58(2):91–6.
  16. Skaper SD, Debetto P, Giusti P. The P2X7 purinergic receptor: From physiology to neurological disorders. FASEB J. 2010;24(2):337–45.
  17. Martin TA, Harrison G, Mansel RE, Jiang WG. The role of P2X7 in cancer: A brief review. J Oncol. 2009;2009:481–87.
  18. Amoroso F, Capece M, Rotondo A, Cangelosi D, Ferracin M, Franceschini A, et al. P2X7 receptor is a cytotoxic receptor selectively expressed on chronic lymphocytic leukemia B cells. Blood. 2015;125(21):346–55.
  19. Adinolfi E, Raffaghello L, Giuliani AL, Cavazzini F, Capece M, Chiozzi P, et al. Expression of P2X7 receptor increases in vivo tumor growth. Cancer Res. 2012;72(12):2957–69.
  20. Di Virgilio F, Vuerich M. Purinergic signaling in the immune system. Auton Neurosci. 2015;191:117–23.
  21. Rissiek B, Haag F, Boyer O, Koch-Nolte F. P2X7 on mouse T cells: One channel, many functions. Front Immunol. 2015;6:204.
  22. Francistiová L, Bianchi C, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer. 2023;23:85–104.
  23. Giuliani AL, Colognesi D, Ricco T, Roncato C, Capece M, Amoroso F, et al. Trophic activity of a naturally occurring truncated isoform of the P2X7 receptor. FASEB J. 2019;33(1):10285–98.
  24. Lees RM, Babon JJ, Brill AL, Hume DA, Stacey KJ, Hamilton JA. P2X7 receptor-deficient mice exhibit impaired osteoclast formation and altered inflammatory responses. J Immunol. 2014;193(2):871–80.
  25. Douguet L, Janho Dit Hreich S, Benzaquen J, Grutter T. Structural basis of P2X7 receptor activation and inhibition. Nat Commun. 2021;12(1):5183.
  26. Ulmann L, Levavasseur F, Avignone E, Peyroutou R, Hirbec H, Audinat E, et al. Involvement of P2X7 receptor in chronic pain models. J Neurosci. 2008;28(44):11106–18.
  27. Sperlágh B, Illes P. P2X7 receptor: An emerging target in CNS diseases. Trends Pharmacol Sci. 2014;35(10):537–47.
  28. Danquah W, Meyer-Schwesinger C, Rissiek B, Csoka B, Scholz K, Schnurr M, et al. Targeting the P2X7 receptor in inflammatory diseases. Expert Opin Ther Targets. 2016;20(4):453–67.
  29. Bhaskaracharya A, Dao-Ung P, Jalilian I, Spildrejorde M, Skarratt KK, Fuller SJ, et al. Pro-inflammatory role of the P2X7 receptor in an animal model of multiple sclerosis. J Neuroinflammation. 2014;11(1):137.
  30. Norenberg W, Gebicke-Haerter PJ, Illes P. Expression of P2X7 receptors on activated microglia in the rat spinal cord following peripheral nerve injury. Brain Res. 2001;926(1–2):206–7.
  31. North RA. Molecular physiology of P2X receptors. Physiol Rev. 2002;82(4):1013–67.
  32. Sluyter R. The P2X7 receptor. Adv Exp Med Biol. 2012;986:83–98.
  33. Gu BJ, Saunders BM, Jursik C, Wiley JS. The P2X7 receptor: A target for new anti-inflammatory therapies. Expert Opin Ther Targets. 2011;15(10):1137–50.
  34. Baroja-Mazo A, Martín-Sánchez F, Gomez AI, Martinez CM, Amores-Iniesta J, Compan V, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014;15(8):738–48.
  35. Keane RW, Dietrich WD, de Rivero Vaccari JP. Inflammasome proteins as biomarkers of multiple sclerosis. Front Neurol. 2018;9:135.
  36. Alves LA, Bezerra RJ, Faria RX, Ferreira LG, da Silva Frutuoso V. Physiological roles and potential therapeutic applications of the P2X7 receptor in inflammation and pain. Mem Inst Oswaldo Cruz. 2013;108(3):341–50.
  37. Nicke A. Homotrimeric complexes formed by the ATP-gated P2X1 receptor. J Biol Chem. 2003;278(13):10419–26.
  38. McGaraughty S, Wismer CT, Zhu CZ, Mikusa J, Honore P, Chu KL, et al. P2X7 receptor antagonism improves sciatic nerve-injury-induced neuropathic pain. Pain. 2007;131(1–2):229–43.
  39. Csóka B, Németh ZH, Szabó I, Davies DL, Vizi ES, Pacher P, et al. Adenosine A2A receptors control IL-10 production by macrophages and attenuate P2X7 receptor-stimulated IL-1β production. J Immunol. 2007;179(12):8280–6.
  40. Hafner M, Koeppl H, Misawa R, Duenas M, Jain A. Quantitative systems pharmacology analysis of ATP-mediated immune signaling in cancer immunotherapy. Cell Rep. 2022;39(9):110900.
  41. Ribeiro DE, Roncalho AL, Glaser T, Ulrich H, Lameu C. The P2X7 receptor in neurodegenerative diseases. Front Cell Neurosci. 2019;13:24.
  42. Illes P, Khan TM, Rubini P. Neuronal P2X7 receptors revisited: Do they really exist? J Neurosci. 2017;37(30):7049–62.
  43. Qu Y, Misaghi S, Newton K, Gilmour LL, Louie S, Cupp JE, et al. P2X7 receptor-stimulated cytokine secretion requires PI3K-dependent NF-κB activation. J Biol Chem. 2007;282(17):12507–18.
  44. Menzies RI, Unwin RJ, Bailey MA. Renal P2 receptors and hypertension. Acta Physiol (Oxf). 2015;213(1):232–41.
  45. Sluyter R, Fuller SJ, Burnstock G. P2X7 receptors in hematopoietic and immune system cells. Purinergic Signal. 2014;10(4):659–64.
  46. Amoroso F, Falzoni S, Adinolfi E, Ferrari D, Di Virgilio F. The P2X7 receptor is a key modulator of the PI3K/GSK-3β axis in microglia. J Neuroinflammation. 2020;17(1):203.
  47. Fang F, Pan J, Xu L, Yu Y, Lu Y, Sun B, et al. Inhibition of P2X7 receptor ameliorates sepsis-induced acute kidney injury in mice. Am J Physiol Renal Physiol. 2021;320(3):F409–21.

Photo
Madhu I Kalasad
Corresponding author

Agm College Of Pharmacy Varur Hubballi.

Photo
Snehal Narayan Mandaokar
Co-author

P. Wadhwani College of Pharmacy Yavatmal.

Photo
Krupa Abhijit Khadakban
Co-author

Sharadchandraa Pawar College Of Pharmacy, Otur Pune.

Photo
Najiya Jakir Tamboli
Co-author

Sharadchandraa Pawar College Of Pharmacy, Otur Pune.

Photo
Nikita Rajesh Gage
Co-author

Abhinav Institute Of Pharmacy Kalyan East.

Photo
Nistha Marwah
Co-author

Dnyan Ganga College Of Pharmacy Thane West.

Madhu I. Kalasad*, Snehal Narayan Mandaokar, Krupa Abhijit Khadakban, Najiya Jakir Tamboli, Nikita Rajesh Gage, Nistha Marwah, Pharmacological Targeting of P2x7 Receptors: Inflammation, Cancer, And Beyond, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 7, 3398-3411. https://doi.org/10.5281/zenodo.16419658

More related articles
A Overview of Proniosome Gel: A Versatile Drug Del...
sushmita chavan, Dr. Sudha Vengurlekar, Sabafarin H. Shaikh, ...
A Review Of Transdermal Patch For Management Of Pa...
Himani sahni, Prabhudatta Panda, Sachin Kumar, ...
Effect Of Active Constituents of Garlic on Cardiov...
Sudarshan Ingale , Pallavi Shelke, Renuka Sukale, Rohini Satdive ...
Determination And Quantification of Alkaloids Content in Tridax Procumbens Leave...
V. Shirisha, A. Ganga Bhavani, A. Varshitha, A. Uday Kumar, K. Guna Priya, K. Satya Deepthi, ...
Formulation Evaluation and Comparative Study of Antitussive Tablet with Marketed...
Swapnali Kapse , Rutuja Kakde , Ashwin Pundkar, Manjusha Markad, Prachi Murkute, Dr. Santosh Payghan...
Related Articles
Social Media and Digital Tools in Adverse Drug Reaction...
Nityapriya Maharana , Jeeban Pradeep Agnihotry , Sai Swagatika Das , Chandrakanta Das , Tushar Kanti...
Formulation And Evaluation of Lipbalm with Natural Ingredients ...
P. Gayatri Devi, CH. Sravanthi, G. Yagnitha, A. Bhargavi, R. Charitha Sri, CH. Bindu Sri, Dr. Sarava...
Virosomes, Hybrid Vehicles For Efficient And Safe Drug Delivery With New Applica...
Prajapati Rahul Vinitbhai , Soni Dhruvi Nileshkumar, Khatiya honey Girdharilal, Rajput Shruti Rakesh...
A View on Prevention of Cervical Cancer by Human Papillomavirus Vaccination...
Gogineni Raja Varsha, Nalajala Bhavana, Katari Akhila, Prathima HN, ...
A Overview of Proniosome Gel: A Versatile Drug Delivery System For Enhanced Ther...
sushmita chavan, Dr. Sudha Vengurlekar, Sabafarin H. Shaikh, ...
More related articles
A Overview of Proniosome Gel: A Versatile Drug Delivery System For Enhanced Ther...
sushmita chavan, Dr. Sudha Vengurlekar, Sabafarin H. Shaikh, ...
A Review Of Transdermal Patch For Management Of Pain...
Himani sahni, Prabhudatta Panda, Sachin Kumar, ...
Effect Of Active Constituents of Garlic on Cardiovascular Disorders...
Sudarshan Ingale , Pallavi Shelke, Renuka Sukale, Rohini Satdive , ...
A Overview of Proniosome Gel: A Versatile Drug Delivery System For Enhanced Ther...
sushmita chavan, Dr. Sudha Vengurlekar, Sabafarin H. Shaikh, ...
A Review Of Transdermal Patch For Management Of Pain...
Himani sahni, Prabhudatta Panda, Sachin Kumar, ...
Effect Of Active Constituents of Garlic on Cardiovascular Disorders...
Sudarshan Ingale , Pallavi Shelke, Renuka Sukale, Rohini Satdive , ...