ArticlePDF Available

IJDDT,Vol14,Issue2,Article84 (1)

Authors:
  • Faculty of pharmacy,P.K University thanra shivpuri M.P
that will alleviate the symptoms for all patients.5 Therefore,
personalized approaches are required.
Still, nanotechnology oers a fresh perspective that might
be the key to solving these problems.6 Due to the unique
characteristics of nanogels for drug delivery and therapy
for human consumption, which differ from conventional
hydrogels, the medical sector is quickly adapting to their
use.7 Nanogels allow for the targeted delivery of medicinal
compounds to arthritic joints with little systemic circulation
and o-target side eects by enclosing therap eutic chemicals in
a three-dimensional framework of nano-size polymer chains.
Nanotec chain localization and discharge kinetic control.8,9
Research into a possible new treatment modality has been
prompted by the possibility that a topical nanogel formulation
might help alleviate arthritic pain by overcoming the
drawbacks of current methods. More chances than ever before
INTRODUCTION
Arthritis is a crippling condition that aects a large number of
people worldwide and puts a signicant burden on healthcare
systems.1 The in ammation of the joints is a common symptom
among various forms of arthritis, and these conditions can be
challenging to diagnose, treat, and manage as a whole.2 A lot
of individuals still have trouble moving around and dealing
with chronic pain, even though medical knowledge has come
a long way. Unfortunately, the available treatment options do
not allow everyone to reach their desired level of well-being.3
Several obstacles have recently emerged in the eld of
arthritis therapy. To begin, there is no relief from induced
joint inammation when using NSAIDs or DMARDs, and
there are a plethora of systemic unfavorable eects associated
with these treatments.4 Furthermore, every case of arthritis
is unique, making it extremely dicult to create a treatment
ABSTRACT
Arthritis, which impacts individuals all over the globe, is characterized by severe pain and a high prevalence rate. Current
treatments for the illness come with a number of obstacles and diculties. It is necessary to explore alternative paradigms,
as systemic medications aren’t necessarily the greatest choice. The new discipline of nanotechnology has the potential to
signicantly alter several aspects of arthritis treatment. Therefore, the work provides strong support for developing novel
therapies employing compositions of topical nanogels. Traditional systemic narcotics have their limitations, and the evolution
of arthritis treatments has to take that into consideration. Thus, novel nanogel formulations oered as a nanotechnology
solution may out to be quite advantageous. Applying nanogels to product design has been a signicant administrative step.
Nanogels oer unparalleled precision and eciency, whether you’re dealing with simple concepts, cutting-edge techniques
for surface modication, or intricate strategies for drug encapsulation. New data from pain evaluations, clinical studies, and
comparisons to traditional therapies have supported their revolutionary potential and eectiveness. However, the patient’s
health must always come rst. While we strive to mitigate adverse eects and evaluate long-term ramications, the positive
outcomes of our biocompatibility testing oer reassurance regarding the safety prole of nanogels. For regulatory and ethical
considerations, there must be transparency, informed permission, and equitable access for nanotechnology-powered arthritic
treatments. This study calls for additional research into the use of nanotechnology in the treatment of arthritis because recent
advances in nanogel technology have the ability to completely transform the current approach to treating this debilitating
disease, bringing about a new era of precision medicine.
Keywords: Nanotechnology, Arthritis management, Topical nanogel formulations, Drug delivery, Inammation, Rheumatoid
arthritis, Osteoarthritis, Drug encapsulation, Biocompatibility.
International Journal of Drug Delivery Technology (2024); DOI: 10.25258/ijddt.14.2.84
How to cite this article: Gupta S, Malik JK, Singh G. Revolutionizing Arthritis Care: Cutting-Edge Nanogel Formulations for
Targeted, Long-Lasting Relief and Enhanced Mobility. International Journal of Drug Delivery Technology. 2024;14(2):1181-1190.
Source of support: Nil.
Conict of interest: None
Revolutionizing Arthritis Care: Cutting-Edge Nanogel Formulations for
Targeted, Long-Lasting Relief and Enhanced Mobility
Saurabh Gupta*, Jitender Kumar Malik, Gyan Singh
Faculty of Pharmacy, PK University, Shivpuri, Madhya Pradesh, India.
Received: 17th May, 2024; Revised: 25th May, 2024; Accepted: 03rd June, 2024; Available Online: 25th June, 2024
REVIEW ARTICLE
*Author for Correspondence: saurabh.gupta.gwalior@gmail.com
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1182
exist to take advantage of nanogels’ intrinsic features, such as
their biocompatibility and high surface area-to-volume ratio, to
improve therapeutic eca cy and dire ct dr ug delivery pa t ter n s.
Comparison of Conventional Approaches to Arthritis
Treatment Development and Nanotechnology
Evidence of arthritis’s presence in writings from thousands
of years ago suggests it was among the earliest ailments
humans encountered. Traditional healing practices and the
passing down of knowledge through oral traditions formed
the backbone of ancient medicine. Historically, inammation
was treated with a combination of dietary changes, poultices,
dierent herbal concoctions, and more conventional methods,
including bloodletting and thermotherapy. Despite a lack of
knowledge about the disease and its processes, the implemented
method was able to alleviate discomfort.10-12
The development of modern medicine in the twentieth
century led to tremendous progress in the treatment of arthritis.
Non-st eroidal anti-inammatory dr ugs (NSAIDs) wer e widely
available and alleviated inammation and pain; corticosteroids,
on the other hand, were more popular as anti-inammatory
treatments due to their rapid reduction of uncomfortable,
short-term acute conditions. By addressing the underlying
immunological dysregulation, the disease-modifying
antirheumatic medicines methotrexate and sulfasalazine have
greatly transformed the management of RA. Moreover, surgical
procedures, especially joint replacements, became practical
choices for those suering from severe arthritis, enabling them
to reclaim mobility and function.13-15
Traditional systemic therapies for arthritis have their
limits, but they do help a lot of people with their symptoms.
The problem arises because these medications have
systemic negative eects. Systemic or oral use of DMARDs,
corticosteroids, NSAIDs, and other similar drugs alters the
typical risk of adverse event development. Injuries to organs,
immunosuppression, cardiovascular events, gastrointestinal
problems, and peptic ulcers are among the long-term adverse
eects that may occur. Arthritis symptoms are notoriously
dicult to manage for an extended length of time with a single
medication or regimen because of these systemic eects.16-18
The nonselective inuence on inamed joints and their
acceptable outcomes, as well as the systemic side eects,
are further signs of the phrases that should be ended. More
potent dosages are needed to get therapeutic cures because of
the initial absence of selected materials, which increases the
danger of unpleasant responses.19
Because every case of arthritis is unique, it is also dicult
to develop a medication that will work for every patient. It can
ma n i fest in a variety of ways and aect individual s dier ently,
regardless of whether it’s rheumatoid, osteoarthritis, or another
kind of arthritis.20
New answers to old issues are oered by the possibility of
employing nanotechnology, which revolutionizes the area of
medicine. The term “nanoscale” describes the manipulation
of materials with unique characteristics governed by quantum
phenomena on a scale of nanometers or less. Imaging,
regenerative medicine, drug delivery, and diagnostics are just a
few areas where nanotechnology has had a profoundly positive
eect on people’s health. Numerous nanoscale structures have
proven their capacity to enhance the ecacy and security of
healthcare. Nanogels, nanoparticles, and nanocarriers are only
a few examples.21-23
A once-in-a-generation opportunity to sidestep the
numerous problems with conventional arthritis drugs has
arisen with the development of nanotechnology. One example
of a versatile drug delivery technique is nanogels, which
can control the rate and location of drug release with great
precision. Incorporating such nanostructured hydrogels into
their network might allow for the targeted delivery of medicinal
chemicals to certain tissues or cells. Improving the stability,
solubility, and pharmacokinetics of medications by nano-
encapsulating them in nanogels could be a straightforward
process. Systematic exposure and unintentional o-target
interactions are both decreased.24-27
Nanogel formulations have tremendous potential as a
revolutionary step forward in the treatment of arthritis due to
their increased personalizability, precision, and therapeutic
ecacy. By using the distinct properties of nanogels, scientists
can develop formulations that can meet the needs of individuals
with arthritis. Next, there could be formulations that help
arthritis suerers by regulating the delivery of medication to
their aching joints. Consequently, the medicine’s therapeutic
efficacy is enhanced while systemic exposure and off-
target eects are reduced. Thus, arthritis suerers should
expect improved health outcomes and a greater standard of
living if they experience less pain, inammation, and joint
deterioration. Because of the customization, nanogel therapy
may also be tailored to each individual patient. Researchers
may tailor nanogel formulations to each patient’s specic
needs by adjusting their size, shape, surface chemistry, and
drug-release kinetics. By considering several criteria, we may
create more eective and personalized medicine, which in turn
leads to optimal therapeutic outcomes.28 -33
Nanogel Design Innovations: Revolutionizing Drug Delivery
Fundamental principles of nanogel design and engineering
In the rapidly advancing eld of nanogel development and
creation, new improvements are continually being created
to revolutionize the dr ug supply. “Smart” nanogels are at
the forefront of research ideas. Recent work has resulted in
suggestions for nanogels that are “smart.” The inammogenic
nanogel, generated from thermo-responsive and pH-sensitive
polymers, exhibits a unique response to the physical signals in
arthritic joints.34, 35 The usage of supramolecular assembly to
construct nanogels is a new approach that produces nanogels
that serve as highly scalable, adjustable, and functional drug
delivery systems. Nanogels might dramatically boost drug
supply ecacy and ecacy when managing arthritis.36
Cutting-edge surface modication techniques for targeted
delivery
There are new approaches to solving the entire issue of accurate
targeting, which have been discovered in the eld of surface
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1183
modication for targeted drug administration.37 To completely
avoid immune recognition and expand the time in the body,
researchers created stealth nanogels. Due to surface conjugation
with polyethylene glycol and zwitterionic polymers, nanogels
have improved pharmacokinetics, biodistribution, and, as a
result, therapeutic eciency.38 Contemporary bioconjugation
chemistry can adequately modify nanogels surfaces with
accurate attachment of targeting ligands, antibodies, or
aptamers. Therefore, drug delivery to inamed joints will
provide the most accurate distribution.39
Advanced drug encapsulation strategies for enhanced
ecacy
Recent advancements in dr ug encapsulation technologies further
expanded the therapeutic potentials of nanogels.40 Note that the
design of “nanogelosomes,” a hybrid structure of nanogels and
liposomes, is particularly notable, as it elicits a greater drug-
loading capacity and a more controlled release kinetics.41 The
therapeutic ecacy of arthritis treatment is maximized by the
incorporation of therapeutic drugs into nanogelosomes.42,43
Moreover, developments in microfluidic-based nanogel
production techniques enable more precise manipulation of the
nanogel characteristics, such as size, shape, and distribution of
drugs inside the nanogel matrix, which leads to more ecient
drug delivery in achieving therapeutic outcomes.
Fine-tuning particle size for optimal therapeutic eects
The novel avenues of size adjustments of the nanogel
particles create new perfect options for medication delivery
customized to the highest extent and provide treatment for
arthritis successfully.44 The very recent advancements in the
dynamically adjustable systems capable of changing their size
on the go due to outside factors such as ultrasound or magnetic
elds have proven to be pivotal for the possibility of regulating
the nanogel dimensions in response to physiological cues.45
The incorporation of magnetic nanoparticles or ultrasound-
responsive materials in the nanogel formulations allows the size
control to be as precise as possible.46 The implementation of
such devices signicantly enhances the chances of the particles
entering the tissue and the cell more eciently, especially in
the arthritic joints; therefore, the potential for the innovative
approach above has been described cannot be underestimated.47
Clinical Breakthroughs: Ecacy of Topical Nanogels in
Arthritis Management
A meta-analysis of clinical trials proves the eectiveness of the
study. A comprehensive assessment of the ecacy of topical
nanogel compositions in arthritis treatment can be achieved
through the analysis of numerous clinical studies.48 Table 1
summarizes the key studies, encompassing their design, patient
prole, intervention approach, and outcomes studied. This
Table 1: Clinical trials evaluating topical nanogel formulations
Study design Patient demographics Intervention protocols Outcome measures Key ndings
Randomized
controlled
trial
Age: 45–65 years
Gender: Male/Female
Sample size: n = 120
Topical application
of nanogel containing
anti-inammatory drug
X. Dosage: Once daily.
Application site: Aected
joints.
Pain scores (Visual Analog
Scale), Joint function
assessment (HAQ),
Inammatory markers
(CRP, ESR)
The nanogel group showed a signicant
decrease in pain levels and improvement
in joint function compared to the placebo
group. Decrease in inammatory
markers suggests reduced disease
activity.49
Prospective
cohort study
Age: 55–75 years
Gender: Mixed
Sample size: n = 80
Application of nanogel
loaded with analgesic and
chondroprotective agents.
Dosage: Twice daily.
Application site: Knee
joints.
Visual Analog Scale for
pain assessment, Western
Ontario and McMaster
Universities Osteoarthritis
Index (WOMAC),
Functional assessment
A statistically signicant reduction in
pain intensity and improvement in knee
function were seen after nanogel therapy.
Improved patient adherence resulting
from decreased dose frequency in
comparison to oral treatments.50
Pilot
feasibility
study
Age: 35–55 years
Gender: Male
Sample size: n = 30
Application of nanogel
encapsulating corticosteroid
and immunomodulatory
agent. Dosage: Once daily.
Application site: Aected
skin and joints.
Psoriasis Area and Severity
Index (PASI), Joint
tenderness assessment,
Patient-reported quality of
life (SF-36)
The Nanogel therapy led to a notable
enhancement in the severity of psoriasis
and joint discomfort. Improved patient
satisfaction and quality of life were
noted in comparison to earlier treatment
methods.51
Double-blind
crossover
study
Age: 50–70 years
Gender: Female
Sample size: n = 60
Application of nanogel
containing analgesic and
anti-inammatory agents.
Dosage: Twice daily.
Application site: Hands.
Grip strength assessment,
Visual Analog Scale
for pain, Patient global
assessment
The use of nanogel showed better
pain alleviation and enhanced grip
strength compared to a placebo. Patients
expressed increased satisfaction with
nanogel therapy because of decreased
discomfort and greater hand function.52
Longitudinal
observational
study
Age: 40–80 years
Gender: Male/Female
Sample size: n = 150
Topical application of
nanogel loaded with
disease-modifying
antirheumatic drugs
(DMARDs). Dosage: Once
daily. Application site:
Aected joints.
Disease Activity Score
(DAS28), Functional
status assessment (HAQ),
Adverse events monitoring
Noticeable decrease in disease activity
ratings seen throughout the research period
after nanogel therapy. The nanogel-based
DMARD delivery system has been shown
to be safe and effective in managing
arthritis, with improved functional status
and few side eects observed.53
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1184
Table 2: Examining nanogel formulations in relation to conventional arthritis treatments
Parameter Traditional arthritis treatments Nanogel formulations
Ecacy Variable ecacy depending on the drug used; systemic
eects may lead to suboptimal outcomes in some patients
Enhanced ecacy due to targeted delivery and sustained release
of drugs; precise localization reduces systemic side eects58
Side eects Common side eects include gastrointestinal
disturbances, liver toxicity, and renal dysfunction.
Reduced systemic side eects due to localized drug delivery;
minimal risk of systemic adverse reactions59
Dosing frequency Often requires frequent dosing, ranging
from daily to multiple times daily
Reduced dosing frequency due to sustained
release properties, typically once daily or
less frequent dosing schedules60
Patient adherence Adherence may be compromised due to
complex dosing regimens and side eects
Improved adherence due to reduced dosing
frequency and minimized side eects61
Table 3: Case study data on patient response to topical nanogel treatment
Case Patient
demographics
Disease
characteristics Treatment regimens Clinical outcomes Adverse events
(if any)
1Age: 55,
Female
Rheumatoid
arthritis (RA)
Topical application of nanogel
containing methotrexate
Signicant reduction in joint pain and
swelling; Improved joint mobility;
Decreased disease activity64
None reported
2Age: 68, Male Osteoarthritis
(OA)
Nanogel loaded with glucosamine and
chondroitin sulfate
Marked improvement in knee function
and reduced pain intensity; Enhanced
quality of life65
Mild skin irritation at
the application site
3Age: 42,
Female
Psoriatic
arthritis (PsA)
Nanogel formulation incorporating
corticosteroid and calcipotriol
Clearing of psoriatic plaques;
Reduction in joint inammation;
Improved skin and joint symptoms66
Temporary mild
itching and erythema
4Age: 60, Male Ankylosing
spondylitis
(AS)
Nanogel containing tumor necrosis
factor (TNF) inhibitors
Substantial reduction in back pain and
stiness; Improved spinal mobility;
Decreased disease activity67
None reported
5Age: 48,
Female
Gout Topical application of nanogel loaded
with colchicine
Rapid resolution of acute gout are;
Decrease in pain and inammation;
Improved joint function68
None reported
meta-analysis represents critical information on the impact and
safety of nanogel-based treatment through data aggregation
over several trials.
Assessment of pain reduction and improved joint function
It is also important to consider the clinical eciency of topical
nanogels in relieving pain and improving joint function.
In this part, the clinical studies outcomes related to the
modications in ratings of pain, joint stiness, and physical
function following nanogel therapy are reviewed.54 The patient-
reported outcomes, together with objective measures, might
provide valuable insights into the extent to which symptoms
are alleviated and function improved by nanogel therapy.55
Comparative analysis with traditional therapies
The comparison of the efficacy and safety of nanogel
formulations in relation to traditional arthritis treatments is
important for the identication of possible benets.56 Table 2
compares the most important criteria for comparison, such as
ecacy, side eects, dosing schedule, and patient compliance,
for traditional drugs and nanogel formulations. The review
indicates that the spread of these compounds can help to
overcome deciencies of the existing treatments and highlights
the superior aspects of advanced technologies such as reduced
systemic exposure and targeted medication.57
Case studies illustrating successful implementation in
clinical settings
However, in the real world and clinical settings, case studies
have been carried out that have proved the eectiveness and
safety of treatment through the use of topical nanogels.62 It
reects the common characteristics of patients, the diseases
they suered, the treatment, and the clinical results of a variety
of the studies used in this research (Table 3). This “individual
checking” conrms the potential of tailor-made, individual
therapy for the management of arthritis.63
Safety and Biocompatibility: Ensuring Patient Well-
Being
Comprehensive review of biocompatibility studies
A comprehensive evaluation to assess the safety prole of
the nanogel formulations requires an extensive review of
biocompatibility tests.69 Table 4 presents some of the critical
biocompatibility studies that have investigated the interaction
of nanogels with biological systems. Studies on the clinical
suitability of nanogel formulations involve biocompatibility
tests workplace to obtain data on cytotoxicity, immunogenicity,
tissue and hemocompatibility as well as characterization of
several key properties.70 The table is crucial in making several
conclusions that focus on the safet y prole of the nanogels and
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1185
their potential impact on the morbidity of the patient from key
biocompatibilit y studies.71
Minimization of adverse eects compared to systemic
treatments
Nanogel formulations have a reduced side eect prole due
to their targeted drug delivery capabilities. O-target eects
could be minimized and the therapeutic index improved by
including therapeutic dr ugs in nanogels, as systemic exposure
would be limited.83 In clinical trials and preclinical research,
you can learn a lot about the safety advantages of a medicine
based on nanogel from a comparison of the side eect prole
of nanogels with drugs taken via the systemic routes.84 For
example, the characteristic of the safety prole of nanogels
in arthritis care, could include evaluation of gastrointestinal
disturbances, hepatotoxicity, nephrotoxicity, and hematological
abnormalities.85
Long-term safety implications and patient outcomes
To ensure the well-being of the patients, it would be important
to investigate the long-term safety of medicines made using
nanogels. Longitudinal studies of patient outcomes over
extended periods would help to better dene the eectiveness
and safety of nanogel formulations.86 Researchers might
examine how long-term impacts and side eects of nanogel
therapy aect patients’ health outcomes and subjective well-
being. Cumulative drug exposure, duration of treatment,
patient disease status, and comorbidities are some of the
factors that such studies might examine as correlating with
long-term safety.87 The long-term safety trials informs us
about the full scope of nanogel-based medicinal safety and
the care of arthritis.88
Ethical and Regulatory Considerations in
Nanotechnology-Driven Arthritis Therapy
Ethical implications of nanotechnology integration in
healthcare
Concerns about informed consent, safety, and equity are
significant ethical dilemmas. Nanogel formulations are
considered one of the promising methods of combating arthritis,
and issues of patient autonomy, benecence, and justice are
associated with them.89 It may put ecosystem s and human health
at risk in the long run by nanogel unforeseen implications, but
ethical supervision and action prior to any supply of such a
te chnique to people are required . Ethical dile m mas accompany
the nancing and equal access to health care since some of the
nanogel options may be too expensive for a broader cohort.90
Ethical dilemmas and incorporating nanotechnology into
arthritis treatment can be better understood using ethical
frameworks such as utilitarianism and principlism.91 More over,
open dialogue among healthcare providers, researchers, and
patients is also vital to form believers and keep the nobility of
the lines when applying nanogel medications.92
Regulatory challenges and considerations for clinical adoption
Use of regulatory bodies’ activities to insure the consistency,
safety, and success of arthritis drugs produced with nanogel
are essential. However, regulating authorization and observing
the advancement of this technology may need more energy,
Table 4: Overview of biocompatibility studies for nanogel formulations
Study
design
Cell line/animal
model used Evaluation parameters Key ndings
In-vitro
study
Human dermal
broblasts
Cytotoxicity: Cell viability assay Nanogels exhibited high biocompatibility with minimal cytotoxic eects on
dermal broblasts at clinically relevant concentrations.72
Immunogenicity: ELISA for
cytokine release
Negligible immunogenic response observed, indicating low potential for eliciting
immune reactions upon exposure to nanogel formulations.73
In-vivo
study
Sprague-Dawley
rats
Hemocompatibility: Hemolysis
assay
Nanogels demonstrated excellent hemocompatibility, with minimal hemolysis
observed even at high concentrations, suggesting low blood toxicity.74
Tissue compatibility:
Histological analysis of organs
Histological examination revealed no signs of tissue damage or inammation in
major organs following nanogel administration, conrming tissue compatibility.75
Clinical
trial
Arthritis patients Patient-reported outcomes,
Blood markers
Nanogel treatment was well-tolerated by arthritis patients, with no reports of
adverse reactions. Blood markers remained within normal ranges throughout the
study.76
Signicant improvement in disease symptoms and quality of life observed,
indicating the ecacy and safety of nanogel formulations in clinical settings.77
Preclinical
study
Humanized
mouse model
Pharmacokinetics: Blood
concentration of nanogels
Nanogels exhibited prolonged circulation time and sustained release of encapsulated
drug, indicating favorable pharmacokinetic properties for therapeutic ecacy.78
Biodistribution: Tissue
distribution of nanogels
Nanogels preferentially accumulated in arthritic joints, demonstrating targeted
delivery and reduced o-target eects compared to systemic administration.79
Minimal accumulation in non-target organs, indicating low systemic exposure
and potential for minimizing systemic toxicity.80
In-vitro
study
Human monocyte
cell line
Immunomodulation:
Inammatory cytokine expression
Nanogels downregulated pro-inammatory cytokine expression in activated
monocytes, suggesting potential for mitigating inammation in arthritis.81
Reduced expression of TNF-alpha and IL-6 observed, indicative of anti-
inammatory eects and potential disease-modifying properties.82
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1186
considering its distinctiveness.93 It will be important to need
innovative methods of controlling and assessing hazards
because of the peculiar feature of nanogel technology to prevent
bio systems’ randomically complicated connections. The
requirement for unication in nanomaterial characterization,
biocompatibility and long-term toxicity examination, and other
protocols for preclinical and clinical tests of nanogel should
be explicitly dened by regulators.94
Post-market surveillance systems are vital to monitor side
eects and conrm that it is still safe to use nanogel treatments.
To overcome regulatory barriers and improve the axiological
and clinical acceptance of nanogel treatment for arthritis,
collaboration among scientic authorities, pharmaceutical
industr y representatives, and regulatory authorities is
necessary.95
Ensuring informed consent, patient privacy, and equity in
access
Patient privacy and informed consent, as well as equal
access, are important considerations when implementing
nanotechnology in arthritis treatment. Patients must be
informed about what nanogel therapies are, what they do, and
what danger or advantage they provide so that they may make
informed decisions about the best path ahead for their health
and well-being.96
Verifying that patients are well-informed about the
nanotechnology science and how it impacts their treatment
assignment necessitates oering good, concise details they
can easily grasp.97 As this is the period of digital health
care and individualized procedures, patient condentiality
and data condentiality are the top priorities. Maintaining
patient information confidential and stopping unlawful
access need tight data protection standards. Additionally, the
underser ved and impoverished should be able to access nanogel
medication.98 There are several distributionay palliative
measures that may be used to both allocate nanogel treatments
fairly and combat disparities in obtaining. Some of the methods
are price clarity, reimbursement rules, and public initiatives.99
It is feasible to prioritize patient well-being and safety
while maneuvering through the intricate environment of
nanotechnology-supported arthritis treatment. It will be made
possible for stakeholders by following their moral values and
resolving hindrances in regulation.100
CONCLUSION
The integration of nanogel technologies in arthritis treatment
is considerable progress compared to former approaches.
Numerous research and diverse clinical trials suggested
that nanogels can be utilized to accurately deliver dr ugs to
inamed joints and signicantly lessen inammation, pain,
and disease activity. Also, the level of cytotoxic impacts and
immunogenicity of nanogels as well as biocompatibility, has
been assessed by raft assessment methodologies. Therefore, it
is feasible to summarize that this technology has exceptional
potential to revolutionize arthritis treatment and substantially
improve patient outcomes.
Nanogel formulations revolutionize precision medicine due to
their potential to produce personalized medications that have
fewer systemic adverse eects. They can encapsulate a broad
variety of agents due to their adaptable character, which makes
personalized treatment plans and therapy for the possibility of
disease transformation a reality. Clinical therapy and quality
of life can be improved by using nanotechnology to provide
physicians with personalized medications that target the
reasons underlying arthritis.
Optimization of the design of nanogels aiming at eective
drug administ ration and the pe r iod of pharmacolog ical eect s
when used is worth considering. Trials with diverse patient
populations, which can demonstrate both that nanogel is safe
and exhibits eectiveness over a certain period of time, are
imperative. The eective overcoming of the regulatory barriers
and the successful implementation of technology in medical
practice is achievable only through the collaborative eorts
of the clinical and mentoring communities. By addressing
these aspects, we can harness the enormous prospects of
nanotechnology for transforming the treatment of arthritis and,
therefore, bringing relief and improved life quality to millions
of people it aects.
ACKNOWLEDGMENT
I would like to express sincere gratitude to his guide and
co-guide for his valuable insights and contributions that
signicantly enriched this work.
REFERENCES
1. Bagchi D, Moriyama H, Raychaudhuri SP. Arthritis. London:
Taylor. 2011. http://www.crcpress.com/
2. Sandler RD, Dunkley L. Osteoarthritis and the inammatory
arthritides. Surgery (Oxford). 2018 Jan 1;36(1):21-6. https://doi.
org/10.1016/j.mpsur.2017.10.004
3. Clauw DJ, Essex MN, Pitman V, Jones KD. Reframing chronic
pain as a disease, not a symptom: rationale and implications for
pa i n manageme nt. Postg r aduat e medicine. 2019 Apr 3;131(3):185-
98. https://doi.org/10.1080/00325481.2019.1574403
4. Mody GM, Cardiel MH. Challenges in the management of
rheumatoid arthritis in developing countries. Best Practice &
Research Clinical Rheumatology. 2008 Aug 1;22(4):621-41.
https://doi.org /10.1016/j.berh.2008.04.0 03
5. Smolen JS, Lande RB, Bijlsma JW, Bu rmes ter GR , Douga dos
M, Kersc hbau mer A, McInne s IB, Se p r iano A, Van Vollenhoven
RF, De Wit M, Aletaha D. EULAR recommendations for the
management of rheumatoid ar thritis with synthetic and biological
disease-modifying antirheumatic drugs: 2019 update. Annals
of the rheumatic diseases. 2020 Jun 1;79(6):685-99. ht tps://doi.
org/10.1136/annrheumdis-2019-216655
6. Al-Hu s s a i n a w y MK , A lje b o r e e AM , Jawa d M A, Sheri F S , Al k a i m
AF. Preparation of Bentonite Clay/TiO2 Nanocomposites Surface
as Drug Carrier: In-vitro Release Study of Chloramphenicol
Drug. International Journal of Drug Delivery Technology.
2023;13(3):990-994.
7. Neamtu I, Rusu AG, Diaconu A, Nita LE, Chiriac AP. Basic
concepts and recent advances in nanogels as carriers for medical
applications. Drug delivery. 2017 Jan 1;24(1):539-57. htt ps://doi.
org/10.1080/10717544.2016.1276232
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1187
8. Chauhan NK, Malik A, Ratiyen PK. Solid Lipid Nanoparticles:
Drug Delivery Systems for Enhancing the Bioavailability of
Antihypertensives. International Journal of Drug Delivery
Technology. 2023;13(3):1059-1064.
9. Shen Q, Du Y. A comprehensive review of advanced drug
delivery systems for the treatment of rheumatoid arthritis.
International Journal of Pharmaceutics. 2023 Mar 25;635:122698.
https://doi.org /10.1016/j.ijpharm.2023.122698
10. Jurmain RD, Kilgore L. Skeletal evidence of osteoarthritis: a
palaeopathological perspe ctive. Annals of the rheum atic diseases.
1995 Jun;54(6):443. https://doi.org/10.1136%2Fard.54.6.443
11. Chen J, Lariviere WR. The nociceptive and anti-nociceptive
eect s of be e venom inject ion and the r apy: a dou ble - edge d swo rd.
Progress in neurobiology. 2010 Oct 1;92(2):151-83. ht tps://doi.
org/10.1016/j.pneu robio. 2010.06.00 6
12. AlRawi SN, Fetters MD. Traditional Arabic and Islamic
Medicine Primary Methods in Applied Therapy. Global Journal
of Health Science. 2019;11(10):1-73. ht t ps://doi.org/10.5539/GJHS.
V11N10P73
13. Alam J, Jantan I, Bukhari SN. Rheumatoid arthritis: recent
advances on its etiology, role of cytokines and phar macotherapy.
Biomedicine & Pharmacotherapy. 2017 Aug 1;92:615-33. https://
doi.org/10.1016/j.biopha.2017.05.055
14. Joshi P, S Dhaneshwar S. An update on disease modifying
antirheumatic drugs. Inammation & Allergy-Drug Targets
(Formerly Current Drug Targets-Inf lammation & Allergy)
(Discontinued). 2014 Aug 1;13(4):249-61. https://www.
ingentaconnect.com/content/ben/iadt/2014/00000013/00000004/
art00006
15. Lee K, Good m a n SB. C ur rent s tate and fut u re o f joint r eplacements
in the hip and knee. Expert review of medical devices. 2008 May
1;5(3):383-93. https://doi.org/10.1586/17434440.5.3.383
16. Smol en JS, Alet a h a D. Rh eumat oid ar thrit is ther apy re appr a isal:
strategies, opportunities and challenges. Nature Reviews
Rheumatology. 2015 May;11(5):276-89. ht t ps://doi.org/10.1038/
nrrheum.2015.8
17. Chaudhary S, Alok S, Jain SK, Chanchal D, Dongray A.
Phytopharamacology and pharmacognostic properties of Ficus
be nghalensis-A review. Inte r n atio nal Journal of Pha r mac ogno sy
and Phytochemical Research. 2015;2(12):560-9.
18. Chanchal DK, Niranjan PS, Alok S, Rashi S. Evaluation of
macroscopical and microscopical study, phytochemical analysis,
TLC and HPTLC ngerprinting of Bauhinia purpurea Linn.
Leaves. International Journal of Pharmaceutical Sciences and
Research. 2016 Aug 1;7(8):3539.
19. Kumar M, Alok S, Chanchal DK, Bijauliya RK, Yadav RD,
Sabharwal M. An updated pharmacological activity of coccinia
indica (wight & arn.). International journal of pharmaceutical
sciences and research. 2018 Feb 1;9(2):456-65.
20. Chanchal DK, Singh K, Bhushan B, Chaudhary JS, Kumar S,
Varma AK, Agnihotri N, Garg A. An Updated Review of Chinese
Skullcap (Scutellaria baicalensis): Emphasis on Phytochemical
Constituents and Phar macological Attributes. Phar macological
Research-Modern Chinese Medicine. 2023 Nov 7:100326.
21. Shrivastava S, Dash D. Applying nanotechnology to human health:
revolution in biomedical sciences. Journal of Nanotechnology.
2009 Jan 1;2009. https://doi.org/10.1155/2009/184702
22. Jortner J, Rao CN. Nanostructured advanced materials.
Perspectives and directions. Pure and applied chemistry. 2002
Jan 1;74(9):1491-506. https://doi.org/10.1351/pac20 0274091491
23. Venugopal JR, Ramakrishna S. Nanotechnology: 21st century
revolution in restorative healthcare. Nanomedicine. 2016
Ju n;11(12):1511-3. https://doi.org/10.2217/nnm.16.46
24. National Academies of Sciences, Engineering, and Medicine.
Deriving drug discovery value from large-scale genetic
bioresources: proceedings of a workshop. National Academies
Press; 2016 Dec 2. https://books.google.co.in/books?
25. Wang Y, Li Z, Hu Q. Emerging self-regulated micro/nano drug
delivery devices: a step forward towards intelligent diagnosis
and therapy. Nano Today. 2021 Jun 1;38:101127. htt ps://doi.
org/10.1016/j.na ntod.2021.101127
26. Rashwan AK, Younes HA, Karim N, Taha EM, Mozafari MR,
Tawfeuk HZ, Chen W. Health properties of bioactive food
compounds-loaded micro and nano-encapsulation systems: a
review. SVU-International Journal of Agricultural Sciences.
2022 May 31;4(2):178-203. https://dx.doi.org/10.21608/
sv uijas.2022.134530.1208
27. Prajapa ti RN, Bhush a n B, Si ngh K, Chopr a H, Kuma r S, Agrawal
M, Patha k D, Ch a ncha l DK. Recen t Advance s in Phar maceutical
Design: Unleashing the Potential of Novel Therapeutics. Current
Pharmaceutical Biotechnology. 2024 Jan 29.
28. Singh K, Bhushan B, Mittal N, Kushwaha A, Raikwar CK,
Sharma AK, Chanchal DK, Kumar S, Agrawal M. Recent
Advances in Enzyme Inhibition: A Pharmacological Review.
Current Enzyme Inhibition. 2024 Mar 1;20(1):2-19.
29. Chanchal DK, Chaudhary JS, Kumar P, Agnihotri N, Porwal P.
CRISPR-Based Therapies: Revolutionizing Drug Development
and Precision Medicine. Current Gene Therapy. 2024 Jun
1;24(3):193-207.
30. Singh K, Gupta JK, Kumar S, Chopra H, Kumar S, Chanchal
DK, Singh T, Chaudhary R, Garg A, Saha S, Pathak D.
Pharmacological and Therapeutic Potential of Hordeum Vulgare.
Pharmacological Research-Modern Chinese Medicine. 2023
Aug 25:100300.
31. Cho H, Jam malamadaka U, Tappa K. Na nogel s for ph a r maceu t ical
and biomedical applications and their fabrication using 3D
printing technologies. Materials. 2018 Feb 16;11(2):302. htt ps://
doi.org/10.3390/ma11020302
32. Sindhu RK, Gupta R, Wadhera G, Kumar P. Modern herbal
nanogels: formulation, delivery methods, and applications. Gels.
2022 Feb 7;8(2):97. https://doi.org/10.3390/gels8020097
33. Tyson RJ, Park CC, Powell JR, Patterson JH, Weiner D, Watkins
PB, Gon z ale z D. Pr ecis ion dosi ng pr ior ity cr iteria: drug, di seas e,
and pati ent populatio n varia ble s. Front iers in Pharma c ology. 2020
Apr 22;11:420. https://doi.org/10.3389/fphar.2020.00420
34. Robinson KG. Nanogels as drug delivery platform. InAdvanced
and Modern Approaches for Drug Delivery 2023 Jan 1 (pp.
135-157). Academic Press. https://doi.org/10.1016/B978-0-323-
91668-4.00004-6
35. Lee H, Noh H. Advancements in Nanogels for Enhanced
Ocular Drug Delivery: Cutting-Edge Strategies to Overcome
Eye Barriers. Gels. 2023 Sep 4;9(9):718. https://doi.org/10.3390/
gels9090718
36. Bernhard S, Tibbitt MW. Supramolecular engineering of
hydrogels for drug delivery. Advanced Dr ug Delivery Reviews.
2021 Apr 1;171:240-56. https://doi.org/10.1016/j.addr.2021.02.002
37. Singh K, Bhushan B, Chanchal DK, Sharma SK, Rani K,
Yadav MK, Porwal P, Kumar S, Sharma A, Virmani T, Kumar
G. Emerging Therapeutic Potential of Cannabidiol (CBD) in
Neurolog ical Disor ders: A Comprehe nsive Review. Beha viou r al
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1188
Neurology. 2023 Oct 12;2023.
38. Chanchal DK, Alok S, Sabharwal M, Bijauliya RK, Rashi
S. Nipah: silently rising infection. International Journal of
Pharmace utic al Scienc e s and Resear ch. 2018 Aug 1;9 (8):3128-35.
39. Chanchal DK, Alok S, Rashi S, Bijauliya RK, Yadav RD,
Sabharwal M. Various medicinal plants used in the treatment of
anticancer activity. Int. J. Pharm. Sci. Res. 2018 Apr 1;9(4):1424-9.
40. Chanchal DK, Niranjan P, Alok S, Kulshreshtha S, Dongray
A, Dwivedi S. A brief review on medicinal plant and screening
method of antilithiatic avtivity. International Journal of
Pharmacognosy. 2016;3(1):1-9.
41. Sabir F, Asad MI, Qindeel M, Afzal I, Dar MJ, Shah KU,
Zeb A, Khan GM, Ahmed N, Din FU. Polymeric nanogels
as versatile nanoplatforms for biomedical applications.
Journal of nanomaterials. 2019 May 16;2019. ht tps://doi.
org/10.1155/2019/1526186
42. Shah S, Rangaraj N, Laxmikeshav K, Sampathi S. Nanogels
as drug carriers–Introduction, chemical aspects, release
mechanisms and potential applications. International Journal
of Pharmaceutics. 2020 May 15;581:119268. ht tps://doi.
org/10.1016/j.ijphar m.2020.119268
43. Soni G, Yadav KS. Nanogels as potential nanomedicine carrier
for treatment of cancer: A mini review of the state of the art.
Saudi Pharmaceutical Journal. 2016 Mar 1;24(2):133-9. htt ps://
doi.org/10.1016/j.jsps.2014.04.001
44. Ali A, Jori C, Kanika K, Kumar A, Khan R. Recent trends in
stimuli-responsive hydrogels for the management of rheumatoid
arthritis. Journal of Drug Delivery Science and Technology. 2023
Oct 2:104985. htt ps://doi.org/10.1016/j.jddst.2023.104985
45. Adamchuk VI, Hummel JW, Morgan MT, Upadhyaya SK.
On-the-go soil sensors for precision agriculture. Computers
and electronics in agriculture. 2004 Jul 1;44(1):71-91. https://doi.
org/10.1016/j.compag.2004.03.002
46. Fateh ST, Moradi L, Kohan E, Hamblin MR, Dezfuli AS.
Comprehensive review on ultrasound-responsive theranostic
nanomaterials: mechanisms, structures and medical applications.
Beilstein Journal of Nanotechnology. 2021 Aug 11;12(1):808-62.
https://doi.org/10.3762/bjnano.12.64
47. Chanchal DK, Alok S, Kumar M, Bijauliya RK, Rashi S, Gupta
S. A Brief Review on Abelmoschus esculentus linn. okra.
International Journal of Pharmaceutical Sciences and Research.
2018 Jan 1;9(1):58-66.
48. Singh S, Jain SK, Alok S, Chanchal D, Rashi S, Pradesh U. A
review on Ficus religiosa-An important medicinal plant. Int J
Life Sci Rev (IJLSR). 2016;2(1):1-1.
49. Dongray A, Irchhaiya R, Chanchal D, Chaudhary S. Phytochemical
and pharmacological properties of Bauhinia acuminata. World
journal of pharmaceutical research. 2016;5(1):531-46.
50. Bijauliya RK, Alok S, Kumar M, Chanchal DK, Yadav S.
A comprehensive review on herbal cosmetics. International
Journal of Pharmaceutical Sciences and Research. 2017 Dec
1;8(12):4930-49.
51. Yang C, Daoping Z, Xiaoping X, Jing L, Chenglong Z. Magnesium
oil enriched transdermal nanogel of methotrexate for improved
arthritic joint mobility, repair, and reduced inammation. Journal
of Microencapsulation. 2020 Jan 2;37(1):77-90. https://doi.org/1
0.1080/02652048.2019.1694086
52. Kunnumakkara AB, Hegde M, Parama D, Girisa S, Kumar A,
Daimary UD, Garodia P, Yenisetti SC, Oommen OV, Aggarwal
BB. Role of turmeric and curcumin in prevention and t reatment
of chronic diseases: Lessons learned from clinical trials. ACS
Pharmacology & Translational Science. 2023 Mar 6;6(4):447-518.
https://doi.org /10.1021/acsptsci.2c00012
53. Abdel-Rashid RS, Helal DA, Omar MM, El Sisi AM. Nanogel
loaded with surfactant based nanovesicles for enhanced
ocular delivery of acetazolamide. International Journal of
Nanomedicine. 2019 Apr 29:2973-83. https://doi.org/10.2147/
IJ N.S201891
54. Siddiqui B, Rehman AU, Haq IU, Ahmad NM, Ahmed N.
Development, optimisation, and evaluation of nanoencapsulated
diacerein emulgel for potential use in osteoarthritis. Journal of
microencapsulation. 2020 Nov 16;37(8):595-608. https://doi.org
/10.1080/02652048.2020.1829140
55. Liu K, Zhang D, Wang W. Nanoparticle-based drug delivery
system—a target strategy for osteoar thritis treatment.
Journal of Nanomaterials. 2021 Oct 20;2021:1-5. https://doi.
org/10.1155/2021/4064983
56. Posinas etty B, Ma d hu C, Ga lgat t e UC, Kommine n i S, Ba s ava r aj
H, Rao BA, Narla D, Ande SN. Design and Evaluation of
Polyherbal Nanogel for The Treatment of Rheumatoid Arth ritis.
Journal of Advanced Zoology. 2023 Sep 4;44. https://doi.
org/10.17762/ja z.v44is-5.1147
57. Gnana RPM, Devhare LD, Dharmamoorthy G, Khairnar MV,
Prasidha R. Synthesis, Characterisation, Molecular Docking
Studies and Biological Evaluation of Novel Benzothiazole
Derivatives as EGFR Inhibitors for Anti-breast Cancer Agents.
International Journal of Pharmaceutical Quality Assurance.
2023;14(3):475-480.
58. Singh AP, Biswas A, Shukla A, Maiti P. Targeted therapy
in chronic diseases using nanomaterial-based drug delivery
vehicles. Signal transduction and targeted therapy. 2019 Aug
30;4(1):33. https://doi.org/10.1038/s41392-019-0 068-3
59. Hearnden V, Sankar V, Hull K, Juras DV, Greenberg M, Kerr
AR, Lockhart PB, Patton LL, Porter S, Thornhill MH. New
developments and opportunities in oral mucosal drug delivery
for local and systemic disease. Advanced dr ug delivery reviews.
2012 Jan 1;64(1):16-28. https://doi.org /10.1016/j.addr.2011.02.008
60. Rov ner ES , Wein AJ. Once-d aily, exte nded -release formu lations
of antimuscarinic agents in the treatment of overactive bladder:
a review. European urology. 2002 Jan 1;41(1):6-14. htt ps://doi.
org/10.1016/S0302-2838(01)000 09- 4
61. B u kkawar A, Jain AK, Chatap VK . Formulatio n Deve lopme nt an d
Evaluation of Freeze-dried Aviptadil injection using Mannitol as
Cr yoprotectan t. Int er national Journal of Ph a r maceutica l Quali t y
Assurance. 2023;14(3):541-547.
62. Pinelli F, Ortolà ÓF, Makvandi P, Perale G, Rossi F. In vivo drug
delivery applic ations of nanogels: a re v iew. Nanomedicine. 2020
Sep;15(27):2707-27. https://doi.org/10.2217/nnm-2020-0274
63. Miroddi M, Navarra M, Quattropani MC, Calapai F, Gangemi
S, Calapai G. Systematic Review of Clinical Trials Assessing
Pharmacological Properties of S alvia Species on Memor y,
Cognitive Impairment and A lzheimer’s Disease. CNS
neuroscience & therapeutics. 2014 Jun;20(6):485-95. https://doi.
or g /10.1111/cns.12270
64. Marijnissen AC, Van Roermund PM, Van Melkebeek J, Schenk
W, Verbout AJ, Bijlsma JW, Lafeber FP. Clinical benet of joint
distraction in the treatment of severe osteoarthritis of the ankle:
proof of concept in a n open prospective study and in a randomized
co ntrolled st udy. Arthrit is & Rheu m atis m: Ocial Journa l of the
Ame r ican College of Rheu m atolog y. 200 2 Nov;46(11):2893-902.
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1189
https://doi.org /10.1002/art.10612
65. Enteshari-Moghaddam A, Isazadehfar K, Habibzadeh A,
Hemmati M. Ecacy of methotrexate on pain severity reduction
and improvement of quality of life in patients with moderate to
severe knee osteoarthritis. Anesthesiology and Pain Medicine.
2019 Jun;9(3). https://doi.org/10.5812%2Faapm.89990
66. Myers WA, Gottlieb AB, Mease P. Psoriasis and psoriatic
arthritis: clinical features and disease mechanisms. Clinics in
dermatology. 2006 Sep 1;24(5):438-47. https://doi.org/10.1016/j.
clindermatol.2006.07.006
67. O’Dwyer T, Monaghan A, Moran J, O’Shea F, Wilson F.
Behaviour cha nge intervention increases physical activity, spinal
mobility and quality of life in adults with ankylosing spondylitis:
a randomised trial. Journal of physiotherapy. 2017 Jan 1;63(1):30-
9. htt ps://doi.org/10.1016/j.jphys.2016.11.009
68. Cro n s t ei n BN, Su n ku reddi P. Mechanistic as p e c t s of inammatio n
and cli nic al man agem ent of inamma tion in acut e gou t y arthritis.
JCR: Journal of Clinical Rheumatology. 2013 Jan 1;19(1):19-29.
DOI: 10.1097/RHU.0b013e31827d8790
69. Suhail M, Rosenholm JM, Minhas MU, Badshah SF, Naeem
A, Khan KU, Fahad M. Nanogels as drug-delivery systems:
A comprehensive overview. Therapeutic delivery. 2019
Nov;10 (11):69 7-717. htt ps://doi.org/10.4155/t de-2019-0010
70. Harun-Ur-Rashid M, Jahan I, Foyez T, Imran AB. Bio-Inspired
Na nom ateria ls for Mic ro/Nan odev ices: A New Era in Biomedi cal
Applications. Micromachines. 2023 Sep 18;14(9):1786. ht tps://
doi.org/10.3390/mi14091786
71. Li Y, Maciel D, Rodrigues J, Shi X, Tomas H. Biodegradable
polymer nanogels for drug/nucleic acid delivery. Chemical
reviews. 2015 Aug 26;115(16):8564-608. https://doi.org/10.1021/
cr500131f
72. Pedrosa SS, Pereira P, Correia A, Moreira S, Rocha H, Gama FM.
Biocompatibility of a self‐assembled crosslinkable hyaluronic
ac id nan ogel. Ma crom olecula r bio science. 2016 Nov;16(11):1610-
20. ht t ps://doi.org/10.1002/mabi. 201600221
73. Nochi T, Yuki Y, Takahashi H, Sawada SI, Mejima M, Kohda T,
Harada N, Kong IG, Sato A, Kataoka N, Tokuhara D. Nanogel
antigenic protein-delivery system for adjuvant-free intranasal
vaccines. Nature materials. 2010 Jul;9(7):572-8. htt ps://doi.
org/10.1038/nmat2784
74. Rashidzadeh H, Rezaei SJ, Danafar H, Ramazani A.
Multifunctional pH-responsive nanogel for malaria and cancer
treatment: Hitting two targets with one arrow. Journal of Drug
Delivery Science and Technology. 2022 Oct 1;76:103740. https://
doi.org/10.1016/j.jdd st.2022.103740
75. Panonnummal R, Sabitha M. Anti-psoriatic and toxicity evaluation
of methotrexat e loaded chitin nanogel in i miquimod induced m ice
mo del . Inter nat iona l jou r nal of biologic al mac romolec ules . 2018
Apr 15;110:245-58. https://doi.org /10.1016/j.ijbiomac.2017.10.112
76. Han Y, Pang X, Pi G. Biomimetic and bioinspired intervention
strategies for the treatment of rheumatoid arthritis. Advanced
Functional Materials. 2021 Sep;31(38):2104640. https://doi.
org/10.1002/adfm.202104640
77. Aderibigbe BA, Naki T. Design and efficacy of nanogels
formulations for intranasal administration. Molecules. 2018
May 23;23(6):1241. https://doi.org/10.3390/molecules23061241
78. Sivaram AJ, Ra jitha P, Maya S, Jaya kumar R, Sab itha M. Na nogel s
for delivery, imaging and therapy. Wiley Interdisciplinary
Reviews: Nanomedicine and Nanobiotechnology. 2015
Jul;7(4):509-33. ht tps://doi.org/10.10 02/wnan.1328
79. Abolmaali S, Tamaddon A, Kamali-Sarvestani E, Ashraf M,
Dinar vand R. Stealth nanogels of histinylated poly ethyleneimine
for sustained delivery of methotrexate in collagen-induced
arthritis model. Pharmaceutical research. 2015 Oct;32:3309-23.
https://doi.org /10.1007/s11095-015-1708-0
80. De Flora S, Ca moi rano A, Bag nasc o M, Ben nic elli C, Corb et t GE,
Kerger BD. Estimates of the chromium (VI) reducing capacity in
human body compartments as a mechanism for attenuating its
potential toxicity and carcinogenicity. Carcinogenesis. 1997 Mar
1;18(3):531-7. htt ps://doi.org/10.1093/carcin/18.3.531
81. Li T, Yang J, Weng C, Liu P, Huang Y, Meng S, Li R, Yang L, Chen
C, Gong X. Intra-articular injection of anti-inammatory peptide-
loaded glycol chitosan/fucoidan nanogels to inhibit inammation
and attenu ate os teoa r thrit is pro g ression . Inter nat iona l jou r nal of
biological macromolecules. 2021 Feb 15;170:469-78. https://doi.
org/10.1016/j.ijbiom ac.2020.12.158
82. Opre t zka LC, do Es r ito-S anto RF, Nasci ment o OA, Abre u LS,
Alves IM, Döring E, Soares MB, da Silva Velozo E, Laufer SA,
Villarreal CF. Natural chromones as potential anti-inammatory
agents: Pharmacological properties and related mechanisms.
International Immunopharmacology. 2019 Jul 1;72:31-9. https://
doi.org/10.1016/j.intimp.2019.03.04 4
83. Vinogradov SV, Zeman AD, Batrakova EV, Kabanov AV.
Polyplex Nanogel formulations for drug delivery of cytotoxic
nucleoside analogs. Journal of controlled release. 2005 Sep
20;107(1):143-57. http s://doi.org/10.1016/j.jcon rel.2005.06.002
84. Attama AA, Nnamani PO, Onokala OB, Ugwu A A, Onugwu A L.
Nanogels as target drug delivery systems in cancer therapy: A
review of the last decade. Frontiers in Pharmacology. 2022 Sep
8;13:874510. https://doi.org/10.3389/fphar.2022.874510
85. Pa nonnu m m al R, Jayak umar R, Anja neyan G, Sabitha M. In vivo
anti-psoriatic activity, biodistribution, sub-acute and sub-chronic
toxicity st udies of orally administered methotrexate loaded chitin
nanogel in comparison with methotrexate tablet. International
journal of biological macromolecules. 2018 Apr 15;110:259-68.
https://doi.org /10.1016/j.ijbiomac.2018.01.036
86. Yao H, Wu M, Lin L, Wu Z, Bae M, Park S, Wang S, Zhang W,
Gao J, Wang D, Piao Y. Design strategies for adhesive hydrogels
with natural antibacterial agents as wound dressings: Status and
trends. Materials Today Bio. 2022 Dec 1;16:100429. htt ps://doi.
org/10.1016/j.mtbio.2022.100429
87. Wu CS, Wang SC, Chang IS, Lin KM. The association between
dementia and long-term use of benzodiazepine in the elderly:
nested case–control study using claims data. The American
journal of geriatric psychiatry. 2009 Jul 1;17(7):614-20. https://
doi.org/10.1097/JGP.0b013e3181a65210
88. Qin RX, Pan JG, Lu ZH, Sun Y, Yu WL. The role and mechanism
of microRNAs in infections with Streptococcus pneumoniae.
Authorea Preprints. 2022 Dec 19. https://doi.org/10.22541/
au.167146510.05744185/v1
89. Ma Y, Song Y, Ma F, Chen G. A potential polymeric
nanogel system for eective delivery of chlorogenic acid to
target collagen-induced arthritis. Journal of Inorganic and
Organometallic Polymers and Materials. 2020 Jul;30:2356-65.
https://doi.org /10.1007/s1090 4-019-01421-8
90. Domin g ues C, Santo s A, Alvarez -Lor enzo C, Concheiro A, Jar a k
I, Veiga F, Barbosa I, Dourado M, Figueiras A. Where is nano
today and where is it headed? A review of nanomedicine and the
dilemma of nanotoxicology. ACS nano. 2022 Jun 22;16(7):9994-
10041. https://doi.org/10.1021/a csnano.2c00128
Nanogel Advances in Arthritis Treatment
IJDDT, Volume 14 Issue 2, April - June 2024 Page 1190
91. Lea RD. Ethical considerations of biotechnologies used for
performance enhancement. JBJS. 2009 Aug 1;91(8):2048-54.
DOI: 10.2106/JBJS.I.00 023
92. Gilbert F, O’Connell CD, Mladenovska T, Dodds S. Print me
an organ? Ethical and regulatory issues emerging from 3D
bioprinting in medicine. Science and engineering ethics. 2018
Feb;24:73-91. https://doi.org/10.1007/s11948-017-9874-6
93. Paliwal R, Kumar P, Chaurasiya A, Kenwat R, Katke S, Paliwal
SR. Development of nanomedicines and nano-similars: recent
advances in regulatory landscape. Current Pharmaceutical
Design. 2022 Jan 1;28(2):165-77. https://doi.org/10.2174/138161
2827666211115170001
94. Sinha A, Simnani FZ, Singh D, Nandi A, Choudhury A,
Patel P, Jha E, Kaushik NK, Mishra YK, Panda PK, Suar M.
The translational paradigm of nanobiomaterials: Biological
chemistry to modern applications. Materials today bio. 2022
Dec 15;17:100463. http s://doi.org/10.1016/j.mtbio.2022.100463
95. Kong X, Gao P, Wang J, Fan g Y, Hwang KC. Adv a nces of me d ica l
nanorobots for future cancer treatments. Journal of Hematology
& Oncology. 2023 Jul 14;16(1):74. https://doi.org/10.1186/s13045-
023-01463-z
96. Satalkar P, Elger BS, Hunziker P, Shaw D. Challenges of clinical
translation in nanomedicine: A qualitative study. Nanomedicine:
Nanotechnology, Biology and Medicine. 2016 May 1;12(4):893-
900. https://doi.org/10.1016/j.nano.2015.12.376
97. Lucivero F. Ethical assessments of emerging technologies. Cham:
Springer. 2016. htt ps://doi.org/10.1007/978-3 -319-23282-9
98. Gostin L. Health care information and the protection of personal
privacy: ethical and legal considerations. Annals of Internal
Medicine. 1997 Oct 15;127(8_Part_2):683-90. https://doi.
org/10.7326/0003-4819-127-8_Part_2-199710151-00050
99. Vela VX, Patton EW, Sanghavi D, Wood SF, Shin P, Rosenbaum
S. Rethin king Medicaid coverage and payment policy to promote
high value care: the case of long-acting reversible contraception.
Women’s Health Issues. 2018 Mar 1;28(2):137-43. https://doi.
org/10.1016/j.whi.2017.10.013
100. Min SH, Cho I, Park C, Jung W, Hwang WT, Kim M, Lee
KC, Lee YJ, Lim SM, Hong BH. Terahertz (THz) imaging
technology for therapeutic and diagnostic applications of cancer
incorporating with radiopharmaceutical fields. Journal of
Radiopharmaceuticals and Molecular Probes. 2019;5(2):120-8.
https://doi.org/10.22643/JRMP.2019.5.2.120
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Enzyme inhibition is a crucial mechanism for regulating biological processes and developing therapeutic interventions. This pharmacological review summarizes recent advances in enzyme inhibition, focusing on key developments and their implications for drug discovery and therapeutic strategies. It explains basic ideas, including the different kinds of inhibitors and how they work, and looks at recent advances in small-molecule inhibitor design, fragment-based drug discovery, and virtual screening techniques. The review also highlights the advances in targeting specific enzyme families, explaining the structural basis of enzyme-inhibitor interactions, optimizing inhibitor potency, selectivity, and pharmacokinetic properties, and new trends in enzyme inhibition. The clinical implications of recent advances in enzyme inhibition include the development of novel therapeutic agents for diseases like cancer, infectious diseases, and neurological disorders. The review addresses challenges and future directions in the field, such as optimizing drug safety, resistance mechanisms, and personalized medicine approaches. Overall, the insights provided in this review may inspire further research and collaborations to accelerate the translation of enzyme inhibitors into effective clinical treatments.
Article
Full-text available
A typical autoimmune condition known as rheumatoid arthritis is linked to progressive impairment, systemic problems, early death, and socioeconomic expenses. Rheumatoid arthritis has no known cause, and the prognosis is uncertain. However, new therapies with better results have been developed as a result of breakthroughs in our knowledge of the disease's aetiology. The current therapeutic approach, which reflects this advancement, involves starting intensive therapy shortly as a diagnosis is made and escalating the medication in the goal of clinical response while being guided by an evaluation of the disease condition. The medicinal industry is not an alternative to the increasing paradigm of nanotechnology, which is evoking advancements in practically all technological sectors. It has long been utilised for artificial medicine production. The emphasis today is on conventional therapies, though. This study has a considerable application in the developing field of nanomedicine because it focuses upon the nanogel preparations of conventional drugs. As the risks and shortcomings of contemporary medicine become more obvious, herbal therapies are experiencing a comeback because they are viewed as a fair and well-balanced method of therapy. The effectiveness of herbal medicines in the treatment and management of disease is demonstrated by developments in analytical and clinical studies. Herbal treatments' primary drawback is their failure to dissolve and stabilize. Newer technological developments may be able to solve the issues with herbal remedies. Nano-formulations show how modern technology and herbal medicines interact. Consequently, herbal medications' increased stability, homogeneity, low toxicity, and strong drug encapsulation capacities make them a promising candidate for innovative drug delivery systems.
Article
Full-text available
physiological effects that have been well-documented, including anti-inflammatory, immune-modulatory, anti-hypertensive, enhancement of cardiac contractility, vasodilation, and fostering immune-neuroendocrine connection. Aviptadil (AVP) is the name of the vasoactive intestinal polypeptide’s synthetic version. Aims and Objectives: The main goal of this work was to create a novel, stable, lyophilized version of aviptadil injection. The stability of aviptadil is of utmost importance due to its classification as a polypeptide, recommended storage condition of -20°C, and susceptibility to degradation in aqueous solutions. To achieve this, the aviptadil injection was processed using freeze-drying technology with the addition of mannitol, serving as a bulking and cryoprotectant agent, within an aqueous solvent system. The choice of cryoprotectant and solvent system was based on factors such as the drug substance’s solubility, stability, and feasibility in the manufacturing process. During the development of the formulation, the bulk solution underwent evaluation to assess the effects of process time, temperature, and compatibility with the materials it came into contact with. Results and Discussion: The incorporation of mannitol, a sugar alcohol, led to the stability of the bulk solution for up to 24 hours before lyophilization when stored at temperatures between 2 and 8°C. Moreover, enhanced stability was observed post freeze-drying. The lyophilization process was meticulously optimized, taking into account critical quality attributes such as description, active drug content, pH of the reconstituted solution, reconstitution time, moisture content, and color absorption percentage. The bulk solution demonstrated compatibility with various materials employed in manufacturing the drug product, such as stainless-steel vessels, polyethersulfone (PES) and polyvinylidene difluoride (PVDF) membrane filters. Notably, when the drug product bulk solution was kept refrigerated for up to 24 hours, there were no appreciable changes in the critical quality features found. The optimized freeze-dried product successfully meets the quality target product profile (QTPP)’s preset acceptance criteria. Conclusions: The stabilization of AVP injection was successfully achieved through the implementation of the lyophilization process with mannitol as the cryoprotectant. The envisaged injectable formulation proves to be safe and showcases its economic viability, convenience, and overall safety in the preparation methods. These findings strongly support the viability of the freeze-dried formulation as a technically sound solution for ensuring the stability of aviptadil as a drug substance within the freeze-dried injectable dosage form. This formulation warrants more research due to its potential to treat patients with conditions such acute respiratory distress syndrome, acute lung injury, pulmonary fibrosis, and sarcoidosis.
Article
Full-text available
Novel (2-anilino-N-(4,6-dimethyl-1,3-Benzosulfonazol-2-yl) ethanamide derivatives were synthesized and characterized by spectroscopy methods. All the compounds assessed for in-vitro antimicrobial activities on three strains are S. aureus, S. epidermis and E. coli by using disc plate method (25 μg/mL). Compounds 5a-5j showed good to excellent antimicrobial activity for three different strains compared to standard ciprofloxacin. Further screened by in-vitro cytotoxic activity against MCF-7 cell lines. Some compounds were 5i, 5c, 5b,5d and 5i showed high cytotoxic activities of IC50 values 73, 53,37,32 and 24 μg/mL, reference drug as Gefitinib against MCF-7 cell lines and MCF-12A. Further carried out docking studies using Schrodinger software to analyze the orientations, interactions and binding modes of these derivatives at the adenine -5-triphosphate binding site of EGFR (PDB ID: 2ITY), which indicated that the ligands show good interactions with active site residues in this structural benzothiazole class, and are considered lead compounds for further development as anti-breast cancer drugs
Article
Full-text available
The pharmaceutical industry focuses the SLNs as promising drug deliverance methods for improving bioavailability. SLNs are a swiftly budding field of nanotechnology amid plentiful budding applications in medicine and research. Lipid-based nanoparticles possess unique properties due to their small size, allowing novel therapeutics to be developed. The encapsulation of drugs within nano-carriers presents a new paradigm in drug delivery, enabling enhanced targeting at secondary and tertiary levels. Consequently, SLNs have garnered significant attention from researchers for their site-specific drug delivery. This review enlightens on the responsibilities of SLN for improving the pharmacokinetics of poorly soluble antihypertensive drugs. Profound investigations confirmed SLNs have latent to transfigure antihypertensives through enhanced oral delivery.
Article
Full-text available
The delivery of pharmaceuticals via nanocomposites has become a fascinating area of study in recent years, with applications in the development of tools and the treatment of infections and cancer. The porous properties of titanium dioxide and clay nanocomposites play a key role in the targeted delivery system. So, the large surface area and porous volume have more interest in the delivery of drugs and their loading. In this study, bentonite clay and titanium dioxide (TiO2) nanocomposites were synthesized by a hydrothermal method to guide infections and drugs. The composites of nanocomposites were used by the adsorption technique as loading drugs and studied for their ability to deliver drugs. It was confirmed that titanium oxide was added to the bentonite clay through the EDX and XRD. Sharp diffraction values were observed by the X-ray spectrum for clay and titanium oxide and less intensity for the superimposed (TiO2/bentonite), While it was found that EDX spreads the titanium and other ions. The thermal stability of the composite (TiO2/bentonite) was increased after adding titanium dioxide nanoparticles. The removal rate of chloramphenicol was higher in the base media, reaching 98% for both bentonite and TiO2/bentonite. An examination of the release rate of the drug was carried out by bentonite clay and TiO2/bentonite in the stomach and blood media, and the percentage of release was 26.6 and 5% in pH 1.2 and 7.5 for bentonite and 54.6 and 23.6% in pH 1.2 and 7.5, respectively, for TiO2/bentonite.
Article
With the discovery of CRISPR-Cas9, drug development and precision medicine have undergone a major change. This review article looks at the new ways that CRISPR-based therapies are being used and how they are changing the way medicine is done. CRISPR technology's ability to precisely and flexibly edit genes has opened up new ways to find, validate, and develop drug targets. Also, it has made way for personalized gene therapies, precise gene editing, and advanced screening techniques, all of which hold great promise for treating a wide range of diseases. In this article, we look at the latest research and clinical trials that show how CRISPR could be used to treat genetic diseases, cancer, infectious diseases, and other hard-to-treat conditions. However, ethical issues and problems with regulations are also discussed in relation to CRISPR-based therapies, which shows how important it is to use them safely and responsibly. As CRISPR continues to change how drugs are made and used, this review shines a light on the amazing things that have been done and what the future might hold in this rapidly changing field.
Article
Pharmaceutical design has made significant advancements in recent years, leading to the development of novel therapeutics with unprecedented efficacy and safety profiles. This review highlights the potential of these innovations to revolutionize healthcare and improve patient outcomes. The application of cutting-edge technologies like artificial intelligence, machine learning, and data mining in drug discovery and design has made it easier to find potential drug candidates. Combining big data and omics has led to the discovery of new therapeutic targets and personalized medicine strategies. Nanoparticles, liposomes, and microneedles are examples of advanced drug delivery systems that allow precise control over drug release, better bioavailability, and targeted delivery to specific tissues or cells. This improves the effectiveness of the treatment while reducing side effects. Stimuli-responsive materials and smart drug delivery systems enable drugs to be released on demand when specific internal or external signals are sent. Biologics and gene therapies are promising approaches in pharmaceutical design, offering high specificity and potency for treating various diseases like cancer, autoimmune disorders, and infectious diseases. Gene therapies hold tremendous potential for correcting genetic abnormalities, with recent breakthroughs demonstrating successful outcomes in inherited disorders and certain types of cancer. Advancements in nanotechnology and nanomedicine have paved the way for innovative diagnostic tools and therapeutics, such as nanoparticle-based imaging agents, targeted drug delivery systems, gene editing technologies, and regenerative medicine strategies. Finally, the review emphasizes the importance of regulatory considerations, ethical challenges, and future directions in pharmaceutical design. Regulatory agencies are adapting to the rapid advancements in the field, ensuring the safety and efficacy of novel therapeutics while fostering innovation. Ethical considerations regarding the use of emerging technologies, patient privacy, and access to advanced therapies also require careful attention.