Pratik Singh1,2, *
1Rungta College of pharmaceutical science
and research Bhilai, Chhattisgarh.
2Formulation Research & Development
–Non-Orals, Sun Pharmaceutical Industries Ltd, Vadodara, 390020, Gujarat, India.
ABSTRACT
The tumor microenvironment (TME)
is a critical determinant of cancer progression and therapeutic resistance.
Recent advances in nanotechnology offer promising strategies to overcome the
challenges posed by the TME, particularly in enhancing drug delivery and improving
treatment efficacy. This review examines the composition and dynamics of the
TME, emphasizing the complex interactions between cancer cells, stromal cells,
immune components, and extracellular matrix factors that contribute to tumor
growth and metastasis. Nanotherapeutics, with their ability to target the TME
specifically, provide a unique opportunity to bypass the barriers that hinder
traditional cancer therapies, such as hypoxia, acidity, and dense extracellular
matrices. By exploring both passive and active targeting mechanisms, this
review highlights the role of nanomaterials, such as liposomes, polymeric
nanoparticles, and metallic nanoparticles, in overcoming these barriers and
improving therapeutic outcomes. It also discusses the integration of nanotechnology
with immunotherapies and other treatment modalities, enhancing immune responses
and modulating the TME. Despite significant progress, challenges remain in the
clinical translation of these nano-therapeutics. These include issues related
to heterogeneity within the TME, resistance mechanisms, and the potential
toxicity of nanomaterials. The review also covers the latest clinical trials,
offering a comprehensive view of the current landscape and future directions
for nano-based cancer therapies. By synthesizing recent research, this review
provides valuable insights into the potential of nanomedicine in
revolutionizing cancer treatment strategies, addressing key challenges, and
improving patient outcomes.
Keywords:
Tumor microenvironment,
Nano-delivery system, Tumor-associated fibroblast, Tumor-associated
angiogenesis, Tumor-associated hypoxia.
1. INTRODUCTION
1.1 OVERVIEW OF CANCER AND TUMOR MICROENVIRONMENT (TME)
Cancer is a multifaceted disease
characterized by uncontrolled cell proliferation, invasion, and metastasis,
posing significant challenges to global health. By 2020, the global cancer
burden had risen to over 19.3 million new cases and 10 million deaths annually,
highlighting the pressing need for innovative therapeutic strategies. Tumor
progression is not solely dependent on malignant cells but is significantly
influenced by the tumor microenvironment (TME)(1). The TME comprises a dynamic and heterogenous ecosystem of cellular
and non-cellular components, including cancer cells, immune cells, fibroblasts,
endothelial cells, extracellular matrix (ECM), and soluble factors like
cytokines and chemokines. These components collectively contribute to tumor
survival, growth, immune evasion, and therapy resistance. Critical
characteristics of the TME include hypoxia, acidic pH, and abnormal
vascularization, which create a hostile yet exploitable niche for therapeutic
intervention. Understanding the complexity of the TME is essential for
developing effective treatments, as it plays a pivotal role in tumorigenesis
and therapeutic resistance(1).
1.2 ROLE OF
NANOTECHNOLOGY IN MODERN CANCER THERAPY
Nanotechnology has revolutionized cancer
therapy by enabling the design of precise, multifunctional, and biocompatible
nanomedicines. Unlike traditional chemotherapeutics, which lack specificity and
often cause systemic toxicity, nanotherapeutics leverage the unique
characteristics of the TME to enhance drug delivery and efficacy. For instance,
nanoparticles can exploit the enhanced permeability and retention (EPR) effect
in tumors, allowing for the preferential accumulation of therapeutic agents
within cancer tissues. Recent advances in nanotechnology include the
development of stimuli-responsive nanocarriers that release drugs in response
to pH, temperature, or redox potential changes typical of the TME(2). Additionally, nanomedicine facilitates combination therapies,
integrating chemotherapy, immunotherapy, and imaging in a single platform.
Liposomes, polymeric nanoparticles, metallic nanoparticles, and lipid-based
carriers such as those used for mRNA vaccines (e.g., lipid nanoparticles in
COVID-19 vaccines) have been adapted for cancer therapy, illustrating the
versatility of nanotechnology. Nanoparticles also address key challenges in
targeting the TME. They can reprogram immune cells to overcome immune suppression,
enhance the delivery of oxygen to hypoxic regions, and degrade ECM barriers to
improve drug penetration. These approaches not only enhance therapeutic
outcomes but also reduce adverse side effects, paving the way for more
effective and personalized cancer treatments(3).
1.3 OBJECTIVES AND SCOPE OF THE REVIEW
This
review aims to provide a comprehensive analysis of the advances and challenges
in utilizing nanotechnology to target the TME. Key objectives include:
- Exploring the Complexities
of the TME: Understanding its role in cancer progression
and resistance mechanisms.
- Reviewing Recent Advances in
Nanotherapeutics: Highlighting innovations in nanomedicine
designed for TME modulation and targeted drug delivery.
- Identifying Challenges and
Limitations: Addressing biological, technical, and
clinical hurdles in the application of nanotechnology to cancer treatment.
- Discussing Future
Directions: Exploring emerging trends, such as
stimuli-responsive nanocarriers, theragnostic, and the integration of
artificial intelligence in nanotherapeutic design(4).
2.
TUMOR MICROENVIRONMENT (TME): COMPOSITION AND DYNAMICS
The tumor microenvironment (TME) is a multifaceted network of
cellular and non-cellular components that interact dynamically to influence
tumor development, progression, and therapeutic outcomes. Understanding these
components and their unique features provides insight into mechanisms of tumor
behavior and reveals novel therapeutic opportunities, particularly in the realm
of nanotechnology(5).
2.1 Cellular Components
of TME
2.1.1 Immune Cells: Immune cells in the TME are dichotomously categorized into
those promoting tumor suppression and those facilitating tumor progression. Key
immune players include:
·
Macrophages:
Tumor-associated macrophages (TAMs) are a hallmark of the TME. TAMs generally
adopt an M2-like phenotype that promotes tissue remodeling, angiogenesis, and
immunosuppression. M1-like macrophages, in contrast, have anti-tumoral
properties, but their activation is suppressed within the TME due to cytokines
like IL-10 and TGF-β
·
Myeloid-Derived
Suppressor Cells (MDSCs): These cells accumulate in the TME
and inhibit T-cell activation and function, further enhancing immune evasion
·
Tumor-Infiltrating
Lymphocytes (TILs): TILs, such as cytotoxic CD8+ T cells, are
often rendered dysfunctional due to an immunosuppressive environment mediated
by regulatory T cells (Tregs) and inhibitory immune checkpoints (e.g.,
PD-1/PD-L1)
2.1.2 Cancer-Associated Fibroblasts
(CAFs): CAFs are central to the stromal
compartment of the TME and play a dual role in supporting tumor growth and
creating barriers to therapy:
·
Pro-Tumoral
Activities: CAFs secrete growth factors such as fibroblast growth
factor (FGF) and vascular endothelial growth factor (VEGF), which support tumor
growth and angiogenesis. They also deposit extracellular matrix (ECM)
components, contributing to physical rigidity and tumor invasion.
·
Resistance
to Therapy: CAFs shield tumor cells from immune attack by
secreting immune-modulatory cytokines and chemokines, such as CXCL12, which
recruit suppressive immune cells.
2.1.3 Endothelial Cells: Endothelial cells are critical in the formation of
tumor-associated vasculature. Tumor vasculature is highly abnormal,
characterized by irregular structure, poor perfusion, and leakiness:
·
Aberrant
Angiogenesis: Driven by VEGF, the tumor vasculature
supports rapid tumor growth but is often non-functional, leading to hypoxia and
inefficient drug delivery.
·
Therapeutic
Target: Nanoparticles designed to target endothelial markers
(e.g., VEGF receptors) or normalize vasculature have shown promise in
preclinical models.
2.2
Non-Cellular Components of TME
2.2.1 Extracellular Matrix (ECM): The ECM provides structural and biochemical support to tumor
cells, with significant alterations observed in the TME:
·
Composition: Tumors
exhibit increased deposition of collagen, fibronectin, and proteoglycans, which
create a stiff matrix conducive to invasion and metastasis.
·
Barriers
to Therapy: The dense ECM impedes drug penetration, a challenge
being addressed by ECM-degrading nanoparticles
2.2.2 Cytokines and Growth Factors: Cytokines mediate crosstalk between tumor cells and stromal
cells:
·
Pro-Inflammatory
Cytokines: IL-6 and TNF-α drive chronic inflammation, which
promotes tumor growth.
·
TGF-β: A
dual-function cytokine that suppresses early tumor growth but promotes invasion
and immune evasion in later stages
2.2.3 Tumor pH: The TME exhibits extracellular acidification due to aerobic
glycolysis (Warburg effect):
·
Acidification
Impact: Low pH impairs immune cell function and enhances
matrix degradation, promoting metastasis.
·
Therapeutic
Potential: pH-sensitive nanocarriers are being developed to
deliver drugs selectively in acidic tumor environments
2.3
Unique Features of the TME
2.3.1 Hypoxia: Hypoxia results from inadequate oxygen supply due to abnormal
tumor vasculature:
·
Hypoxia-Induced
Pathways: Stabilization of hypoxia-inducible factor-1α (HIF-1α)
drives angiogenesis, metabolic adaptation, and epithelial-mesenchymal
transition (EMT).
·
Therapeutic
Strategies: Oxygen-releasing nanoparticles and hypoxia-activated
prodrugs are being explored to counteract hypoxia
2.3.2 Reactive Oxygen Species (ROS): Elevated ROS levels in the TME promote oxidative stress,
which contributes to genomic instability and tumor progression:
·
Therapeutic
Exploitation: Nanoparticles designed to modulate ROS
levels are emerging as effective tools for inducing cancer cell apoptosis
2.4
Interactions Between TME and Tumor Progression: The TME
influences tumor progression through intricate feedback loops:
- Immune Evasion:
Immunosuppressive cells and cytokines allow tumors to escape immune
surveillance.
- Metastasis:
ECM remodeling and hypoxia facilitate tumor cell detachment and invasion.
- Resistance to Therapy:
The dense ECM and abnormal vasculature limit drug penetration, while
hypoxia and acidity contribute to resistance mechanisms(6).
3. NANOTECHNOLOGY IN CANCER TREATMENT
3.1 Evolution of
Nanomedicine: Key Milestones and Applications Nanotechnology in
cancer treatment has come a long way, with the first key development being the
formulation of liposomal doxorubicin (Doxil®) in the 1990s, which was one of
the first nanomedicines approved by the FDA. This liposomal formulation
utilizes the enhanced permeability and retention (EPR) effect,
a phenomenon where nanoparticles accumulate in tumor tissues due to their
abnormal vasculature and impaired lymphatic drainage. This paved the way for
further advancements in using nanotechnology to improve drug delivery, reduce
toxicity, and enhance the specificity of cancer treatments. As of 2023,
numerous nanoparticle-based therapies have been developed and
clinically tested, ranging from polymeric micelles to metallic
nanoparticles. The development of lipid nanoparticles (LNPs),
especially in the context of mRNA delivery for cancer immunotherapy, has
emerged as a significant milestone. For instance, LNP-based mRNA cancer
vaccines are a promising area of research, inspired by the success of
COVID-19 vaccines (Pfizer-BioNTech, Moderna), which used lipid nanoparticles
for effective mRNA delivery. Additionally, nanotheranostics—the combination of diagnostics and therapeutics
in a single nanoparticle—has gained momentum. Recent innovations focus on
creating nanoparticles capable of both drug delivery and real-time imaging,
using modalities such as magnetic
resonance imaging (MRI) and positron
emission tomography (PET). These "smart" nanocarriers allow
precise tracking of the therapeutic process, enhancing the clinical efficacy of
cancer therapies(7).
3.2
Advantages Over Conventional Therapies: Nanotechnology offers
several advantages over traditional cancer treatments:
·
Targeting: Nanoparticles can be engineered to actively target specific cancer
cells or the tumor microenvironment (TME), reducing off-target effects. This is
achieved by functionalizing the nanoparticle surface with targeting ligands such as
antibodies, peptides, or aptamers, which bind specifically to overexpressed receptors on
cancer cells or tumor vasculature. For example, HER2-targeted liposomes have
shown promising results in breast cancer therapy.
·
Enhanced Penetration: Nanoparticles are small enough to navigate the tumor vasculature and reach
tumor sites more efficiently than conventional therapies. Their small size
allows them to diffuse into the tumor interstitial space and penetrate deep
into the tumor, overcoming physical barriers like the extracellular matrix (ECM) and high interstitial fluid pressure. For instance, polymeric micelles can encapsulate hydrophobic drugs like paclitaxel, which traditionally
suffer from poor solubility and bioavailability.
·
Reduced Toxicity: One of
the major drawbacks of conventional chemotherapy is its toxicity to healthy
tissues. Nanoparticles, particularly those with a lipid or polymeric structure,
can encapsulate drugs and control the
release to minimize damage to normal tissues. For instance, liposomal formulations have shown
reduced systemic toxicity compared to free drugs, allowing higher doses to be
delivered directly to the tumor site. Furthermore, the extended
circulation time of nanoparticles—achieved through surface
modifications like PEGylation—increases their time in the
bloodstream, allowing for sustained drug release and reducing the frequency of
drug administration(8).
3.3 Types of
Nanotherapeutics
a. Liposomes
Liposomes are spherical lipid-based nanoparticles that have
been widely used for cancer treatment. Liposomes encapsulate hydrophilic drugs
in their aqueous core and hydrophobic drugs in their lipid bilayer, offering an
excellent delivery vehicle for a range of anticancer agents. Doxil®, the first FDA-approved
liposomal formulation of doxorubicin, showed better efficacy and fewer side effects than conventional
doxorubicin.
Recent studies have improved liposome
formulations, incorporating pH-sensitive liposomes that can release their payload in response to the acidic pH of the
tumor microenvironment. Liposome-functionalized
nanoparticles targeting specific receptors,
such as folate receptors or EGFR, have also been developed to enhance selectivity and therapeutic
outcomes(9).
b.
Polymeric Micelles
Polymeric
micelles are self-assembled nanoparticles made of amphiphilic block copolymers.
These micelles have a hydrophobic core that encapsulates poorly soluble
anticancer drugs and a hydrophilic shell that provides stability in the
bloodstream. One of the main advantages of polymeric micelles is their ability
to improve the solubility and bioavailability of hydrophobic drugs,
such as paclitaxel, which is
otherwise poorly soluble in water.
Recent
innovations include the development of stimuli-responsive
polymeric micelles, which release their cargo in response to specific
triggers like pH, temperature, or enzymatic cleavage. For example, pH-sensitive micelles release their
contents in the acidic environment of tumors, providing controlled and targeted
drug release(3).
c.
Metallic Nanoparticles
Metallic
nanoparticles, such as gold
nanoparticles (AuNPs), have gained attention for their dual therapeutic
and imaging capabilities. Gold
nanoparticles can be used for photothermal
therapy (PTT), where they are localized at the tumor site and heated
with near-infrared (NIR) light, causing localized tumor destruction.
Additionally, they serve as effective contrast
agents in CT and MRI imaging,
providing real-time monitoring of treatment responses.
Other
metallic nanoparticles, such as iron
oxide nanoparticles, are used for magnetic resonance imaging (MRI) and magnetic hyperthermia—a technique where the nanoparticles generate
heat when exposed to an alternating magnetic field, leading to localized tumor
destruction(9).
Despite
their versatility, challenges remain in ensuring the biocompatibility and long-term
stability of metallic nanoparticles in clinical applications.
d. Lipid
Nanoparticles and mRNA Carriers
Lipid
nanoparticles (LNPs) have become central to the delivery of nucleic acids, such as siRNA, mRNA, and CRISPR/Cas9
systems, for cancer therapy. These LNPs protect the genetic material
from degradation and facilitate its cellular
uptake, ensuring efficient gene
silencing or expression. LNPs are widely recognized for their role in
the successful delivery of mRNA
vaccines (such as the Pfizer-BioNTech and Moderna COVID-19 vaccines),
and this technology is now being repurposed for cancer immunotherapies. mRNA vaccines for cancer, designed to
trigger the immune system to target and destroy cancer cells, are currently
being investigated in clinical trials LNPs are also capable of
co-delivering immune modulators like checkpoint
inhibitors or cytokines, which enhance anti-tumor
immune responses. The combination of mRNA-based therapies with LNPs
holds significant potential for personalized cancer vaccines
and gene editing therapies(10).
Table 1 tumor microenvironment governed
conventional nano-chemotherapeutics targeted for tumor therapy.
4.
NANO-THERAPEUTICS AND TUMOR MICROENVIRONMENT MODULATION
The tumor
microenvironment (TME) is a complex and dynamic ecosystem that consists of not
only cancer cells but also stromal cells, extracellular matrix (ECM), blood
vessels, immune cells, and a variety of soluble factors like cytokines and
growth factors. These components interact in ways that facilitate tumor growth,
metastasis, and resistance to therapies. In recent years, nanotherapeutics have
emerged as a promising approach to target and modulate the TME, offering novel
strategies to improve drug delivery, enhance therapeutic efficacy, and overcome
barriers to effective cancer treatment. Below, we explore the key strategies
used to target and modulate the TME using nanomedicine, based on recent
research and reviews in the field(11).
4.1
Passive Targeting via Enhanced Permeability and Retention (EPR)(12).
The Enhanced
Permeability and Retention (EPR) effect is a
hallmark of many solid tumors and one of the most widely utilized strategies in
cancer nanomedicine. Tumors often possess leaky blood vessels and impaired
lymphatic drainage due to abnormal angiogenesis, which facilitates the passive
accumulation of nanoparticles in the tumor tissue. The EPR effect is based on
the larger pore sizes in tumor vasculature, which allow nanoparticles (usually
in the range of 10–200 nm) to extravasate from the blood circulation into the
tumor interstitium. Furthermore, poor lymphatic drainage prevents efficient
clearance of these nanoparticles, leading to their retention within the tumor
site. However, while EPR has shown promise in preclinical and early clinical
studies, its efficacy is not universal across all tumor types. Variability in
the extent of vascular permeability, the physical properties of nanoparticles,
and the complexity of the TME often result in heterogeneous distribution and
incomplete penetration of nanoparticles into the tumor mass. To enhance the
efficacy of passive targeting, recent research has focused on optimizing
nanoparticle size, surface properties (e.g., surface charge, hydrophilicity),
and the use of nanomaterials that respond to specific TME features such as pH
and oxidative stress(7).
4.2
Active Targeting with Ligands and Antibodies
Active targeting involves the
functionalization of nanoparticles with specific ligands, antibodies, or
aptamers that bind to overexpressed receptors on cancer cells or the tumor
vasculature. Unlike passive targeting, which relies on physical characteristics
of the tumor, active targeting requires the nanoparticle to actively interact
with the target receptor to initiate internalization. Recent studies have
explored a wide range of targeting ligands such as monoclonal antibodies,
peptides, and small molecules that specifically recognize tumor-associated
antigens like HER2, EGFR, folate receptor, and integrins. One major advantage
of active targeting is its ability to increase specificity, reducing off-target
effects and toxicity to normal tissues. Moreover, combination strategies that
pair active targeting with other therapeutic modalities (such as chemotherapy
or gene therapy) are being investigated to improve tumor eradication. However,
a challenge remains in ensuring that nanoparticles can overcome the dense
stromal barriers in some tumors and achieve effective cellular uptake(13).
4.3
Modulation of TME Features
Recent
nanotherapeutic strategies focus on modulating the TME itself, rather than
solely targeting cancer cells. These approaches seek to create a more favorable
environment for treatment efficacy. The modulation of specific TME features,
including hypoxia, acidic pH, and immune suppression, has gained significant
attention in the design of nanoparticles that can alter the tumor's biology.
4.4
Hypoxia Alleviation
Hypoxia,
a common feature of solid tumors, plays a pivotal role in tumor progression and
resistance to therapies, including radiation and chemotherapy. Tumor hypoxia
results from inadequate blood supply due to abnormal vasculature. Hypoxic
regions often contain cancer cells that are more aggressive, invasive, and
resistant to conventional therapies. Nanoparticles that can alleviate hypoxia,
either by delivering oxygen or by modifying the oxygen delivery pathways, have
shown promise in preclinical models. Nanoparticles designed to deliver oxygen
or to act as oxygen carriers can help overcome hypoxia-induced resistance,
improve the efficacy of treatments like radiation therapy, and enhance the
uptake of other therapeutic agents
4.5
pH-Responsive Nanocarriers
The
acidic pH of the TME (often below 6.5) is another distinguishing feature that
offers an opportunity for targeted drug delivery. The pH-sensitive behavior of
nanoparticles is being harnessed to design drug delivery systems that are
stable in the bloodstream but undergo structural changes or drug release when
they encounter the acidic microenvironment of tumors. These pH-responsive
nanocarriers can be functionalized with acid-labile linkers or polymers, such
as poly (ethylene glycol) (PEG), that degrade or release their payload when
exposed to acidic conditions. This strategy allows for the controlled release
of therapeutics specifically within the tumor, reducing side effects and
improving the overall therapeutic index(14).
4.6
Immune Modulation via Nanoparticles
Immune
evasion is a major challenge in cancer treatment, and many tumors develop
immune-suppressive TME characteristics, such as the recruitment of regulatory T
cells (Tregs) and myeloid-derived suppressor cells (MDSCs), which hinder the
anti-tumor immune response. Nanoparticles are being engineered to modulate the
immune landscape of tumors, either by promoting anti-tumor immunity or by
reprogramming immune cells. For instance, nanoparticles can be used to deliver
immune checkpoint inhibitors (e.g., PD-1/PD-L1 inhibitors), adjuvants, or
cytokines directly to the tumor site, enhancing immune activation.
Recent
innovations in nano-immunotherapy include the use of nanocarriers to deliver
STING (Stimulator of Interferon Genes) agonists to "cold" tumors,
thus converting them into "hot" tumors that are more responsive to
immune checkpoint blockade. By boosting the innate immune response, such
strategies have the potential to overcome immune suppression and improve the
efficacy of immunotherapies(15)(16).
4.7
Combination Therapies: Nanomedicine with Chemotherapy, Immunotherapy, and
Radiotherapy
Combining
nanomedicine with traditional therapies such as chemotherapy, immunotherapy,
and radiotherapy is emerging as a promising strategy to overcome the
limitations of each approach when used in isolation. For instance,
nanoparticles can serve as carriers for chemotherapeutic agents, enhancing
their solubility, stability, and bioavailability, while also enabling
controlled and targeted delivery to the tumor site. This can reduce systemic
toxicity and improve the therapeutic efficacy of chemotherapy. Nanoparticles
are also being investigated for their ability to sensitize tumors to radiation
therapy. This includes the use of radiosensitizing nanoparticles that
accumulate in tumor cells and increase the local radiation dose, thereby
enhancing the effects of radiotherapy. Additionally, combining nanomedicine
with immunotherapies, such as immune checkpoint inhibitors or cancer vaccines,
has shown potential for synergistically improving anti-tumor immune responses(15).
5. RECENT ADVANCES IN NANO-THERAPEUTICS(17)
In the field of cancer treatment,
recent advances in nanotechnology have significantly improved the potential of
therapeutic approaches, specifically through the development of novel
nanocarriers, stimuli-responsive materials, imaging, and theranostic integration.
These innovations aim to better target the tumor microenvironment (TME),
enhance drug delivery, and reduce side effects. Here, we explore some of the
most promising recent advancements in nano-therapeutics:
5.1
Innovations in TME-Specific Nanocarriers
The tumor microenvironment (TME) presents
significant challenges to effective cancer treatment due to its unique and
complex properties, including altered pH, hypoxia, and abnormal vasculature.
Recent innovations in TME-specific nanocarriers have been developed to exploit
these unique features of the TME for enhanced drug delivery.
1.
pH-Responsive
Nanocarriers: Tumors are typically more acidic
than normal tissues due to increased metabolic activity and insufficient blood
supply. Exploiting this acidic environment, pH-sensitive nanoparticles have
been developed to release drugs specifically in the tumor site. These
nanocarriers are often composed of polymeric materials such as poly(ethylene
glycol) (PEG) or poly(lactic-co-glycolic acid) (PLGA), which undergo
conformational changes at low pH, leading to drug release. Recent studies have
shown that nanoparticles like DOX-loaded liposomes and pH-sensitive micelles
can improve the specificity and effectiveness of chemotherapy while minimizing
systemic toxicity.
2.
Targeting Tumor
Vasculature and the Extracellular Matrix (ECM):
Abnormalities in tumor vasculature create gaps between endothelial cells, which
allow for passive targeting through the Enhanced Permeability and Retention
(EPR) effect. Recent advancements focus on improving this effect by designing
nanoparticles that can target specific markers expressed by the endothelial
cells of the tumor blood vessels, such as integrins and glycoproteins.
Nanoparticles that target the ECM components like fibronectin and collagen have
also shown promise in enhancing tumor penetration and retention.
3.
Immune-Microenvironment
Modulation: Another innovative approach
includes the use of nanoparticles that can alter the immune response in the
TME. For example, nanoparticles can deliver immunomodulatory agents such as
cytokines or checkpoint inhibitors directly to the tumor site, thus promoting
anti-tumor immunity. Nanoparticles like exosome-based systems or dendritic
cell-based nanovaccines have been explored for their ability to activate immune
cells, further enhancing their potential for cancer immunotherapy.
5.2
Stimuli-Responsive Nanomaterials for Targeted Delivery(17)
Stimuli-responsive nanomaterials, which
respond to internal (e.g., pH, temperature, and enzymatic activity) or external
(e.g., light and magnetic fields) stimuli, represent a rapidly evolving class
of drug delivery systems for cancer therapy. These nanomaterials enable precise
control over the release of therapeutic agents, ensuring that drugs are
delivered only to the tumor site or at specific times during treatment.
1.
Thermoresponsive
Nanomaterials: These materials can be
triggered by changes in temperature, such as those occurring in the TME due to
inflammation or external heating methods (e.g., hyperthermia). Thermoresponsive
polymers, such as poly(N-isopropylacrylamide) (PNIPAAm), have been widely
studied for their ability to transition between hydrophilic and hydrophobic
states in response to temperature changes, thus facilitating controlled drug
release in response to localized heat.
2.
Enzyme-Responsive
Nanocarriers: Tumors exhibit higher levels of
specific enzymes like matrix metalloproteinases (MMPs) or cathepsins, which can
be exploited for drug release. Recent research has focused on developing
nanocarriers that are coated with substrates susceptible to cleavage by these
enzymes. This approach enhances the specificity of drug release in the tumor
tissue, limiting systemic exposure and side effects. For instance,
nanoparticles conjugated with peptide sequences that are cleavable by MMPs have
shown promising results in the delivery of anticancer drugs.
3.
Light and Magnetic
Field-Triggered Nanomaterials: External
stimuli such as near-infrared (NIR) light or magnetic fields offer non-invasive
and spatially controlled methods for triggering drug release. NIR light has
been particularly useful in photo-therapeutic systems due to its deep tissue
penetration, which can trigger the release of drugs from gold nanoparticles,
quantum dots, or polymeric micelles loaded with chemotherapy agents.
Additionally, magnetic nanoparticles can be manipulated by external magnetic
fields to enhance drug delivery and provide a controlled release in response to
specific tumor location.
5.3
Integration of Imaging and Theranostics(15)
The integration of diagnostic imaging and
therapeutic modalities into a single platform, known as theranostics, is a
major breakthrough in cancer treatment. Nanoparticles can be engineered to
carry both therapeutic agents and imaging probes, enabling real-time monitoring
of treatment efficacy while simultaneously delivering therapeutic payloads.
1.
Nanoparticles for
Imaging: Nanoparticles such as
superparamagnetic iron oxide nanoparticles (SPIONs), gold nanoparticles, and
quantum dots are commonly used in molecular imaging techniques like magnetic
resonance imaging (MRI), computed tomography (CT), and fluorescence imaging.
These nanoparticles enhance the sensitivity and resolution of imaging methods,
allowing for better visualization of tumors and tracking of nanoparticle
biodistribution.
2.
Theranostic
Nanoparticles: Nanoparticles are being
designed to combine therapeutic action with diagnostic capabilities. For
example, liposomal formulations containing both chemotherapy agents and
contrast agents have been developed for MRI-based tumor imaging and targeted
therapy. This dual-action approach not only helps in tracking the therapy's
effectiveness but also enables real-time adjustments in treatment regimens
based on imaging feedback.
3.
Biomarker-Targeted
Theranostic Nanomaterials: Recent developments
have also focused on engineering nanoparticles that target specific tumor
biomarkers, allowing for personalized medicine approaches. These nanoparticles
can be loaded with drugs and coupled with diagnostic agents that recognize biomarkers,
providing a precise and non-invasive method to monitor therapeutic response.
This integration improves the efficacy of treatments while minimizing
unnecessary exposure to healthy tissues(18).
5.4
Advances in Biodegradable and Biocompatible Nanomaterials
The biocompatibility and biodegradability
of nanomaterials are critical factors in their clinical translation, as they
influence both safety and therapeutic efficiency. Recent advancements have
focused on developing nanocarriers that degrade into non-toxic byproducts and
exhibit minimal immune reactions.
1.
Biodegradable Polymers: Biodegradable polymers such as PLGA, PEG, and polycaprolactone
(PCL) have been widely used in the development of nanocarriers for cancer
therapy. These materials can be engineered to degrade in a controlled manner,
releasing the drug over a specified period and ultimately breaking down into
non-toxic components that are easily eliminated from the body.
2.
Natural Polymer-Based
Nanomaterials: The use of natural polymers
like chitosan, alginate, and hyaluronic acid in nanomedicine has gained
attention due to their inherent biocompatibility and ability to target specific
receptors overexpressed in cancer cells. For instance, hyaluronic acid-based
nanoparticles can target CD44 receptors, which are highly expressed in many
cancer cells, providing a targeted drug delivery approach with reduced side
effects(19).
3.
Safety and Immune
Response: Advances in understanding the
interactions between nanomaterials and the immune system have led to the
development of nanoparticles that minimize immune system activation. For
example, PEGylation of nanoparticles (the attachment of polyethylene glycol) is
a well-known method to reduce immune recognition and increase the circulation
time of nanoparticles in the bloodstream(20).
6. CHALLENGES IN NANO-THERAPEUTICS FOR CANCER
Nanotherapeutics have shown significant
promise in overcoming the limitations of traditional cancer treatments, but
their clinical translation faces a series of challenges, especially due to the
complex nature of the tumor microenvironment (TME) and the intricacies of
nanomaterial interactions with biological systems. Below are some of the most
pressing challenges identified in recent research.
6.1
Heterogeneity of Tumor Microenvironment (TME) and Its Implications
One of the most significant barriers to
the effectiveness of nano-therapeutics in cancer treatment is the inherent
heterogeneity of the TME. The TME is composed of a variety of cells (e.g.,
cancer cells, stromal cells, immune cells) and extracellular components (e.g.,
extracellular matrix, blood vessels, cytokines) that exhibit significant
spatial and temporal variability. This heterogeneity leads to a range of
difficulties, including inconsistent drug distribution, inefficient targeting,
and variations in response to treatment across different tumor types and even
within different areas of the same tumor. Several studies have shown that the
tumor vasculature, often irregular and leaky, can hinder the efficient delivery
of nanomaterials, with nanoparticles preferentially accumulating at the
periphery rather than uniformly throughout the tumor mass. This phenomenon,
known as the "enhanced permeability and retention" (EPR) effect, is
limited by the chaotic nature of tumor blood vessels, which often result in poor
penetration of nanomedicines into deeper tumor regions. Additionally, some
tumors possess a dense stromal layer, which further restricts nanoparticle
diffusion, reducing the overall efficacy of treatment. Moreover, the
variability in immune cells within the TME plays a role in modulating treatment
responses. The TME can be immunosuppressive, particularly in "cold"
tumors that lack sufficient immune cell infiltration, thereby limiting the
effectiveness of immunotherapies and nanoparticle-mediated immune modulation.
To overcome this challenge, ongoing research is focused on developing
nanoparticles capable of not only enhancing drug delivery but also modulating
the immune microenvironment to promote better therapeutic responses(21).
6.2
Nanocarrier Biodegradability and Long-Term Toxicity
Another major challenge in the clinical
application of nanomedicines is ensuring their safe biodegradability and
minimizing long-term toxicity. Many nanocarriers are designed using synthetic
polymers, lipids, or metals that do not naturally degrade in the body, posing a
risk of accumulation in vital organs like the liver, kidneys, and spleen. Over
time, this accumulation could lead to adverse effects such as inflammation,
organ dysfunction, or the development of secondary diseases. Recent
advancements in biodegradable nanocarriers aim to address this issue by
utilizing materials that break down into non-toxic by-products. For example,
polymeric nanoparticles such as those made from poly (lactic-co-glycolic acid)
(PLGA) are designed to degrade after delivering the therapeutic payload.
However, the degradation rate of these nanoparticles must be carefully
controlled to ensure that they do not release toxic metabolites prematurely. Similarly,
lipid-based nanocarriers, such as liposomes and solid lipid nanoparticles, have
been developed with improved biodegradability, but the challenge remains in
balancing the stability of the carrier with its ability to degrade in a timely
manner without inducing adverse effects. Ongoing research into the long-term
biocompatibility of these nanomaterials is essential to ensure that
nanomedicines can be used safely in humans without risking cumulative toxicity.
The development of nanocarriers with controlled release profiles, targeting
specificity, and safer degradation pathways remains a high priority in
nanomedicine research(19).
6.3
Drug Resistance Mediated by TME
The development of drug resistance remains
one of the most significant hurdles in cancer therapy, and the TME plays a
crucial role in this phenomenon. Tumor cells are often able to adapt to
external stressors, including chemotherapy or targeted therapies, through
various mechanisms facilitated by the TME. For example, the acidic and hypoxic
conditions of many tumors can induce resistance by promoting the expression of
drug efflux pumps, altering drug metabolism, or upregulating protective
pathways like autophagy and DNA repair mechanisms. Nanomedicines designed to
target specific molecular pathways within the TME can sometimes exacerbate drug
resistance. For instance, certain nanoparticles may trigger the activation of
protective immune cells or promote the secretion of cytokines that enhance
tumor cell survival. Moreover, the poor penetration of nanoparticles into
deeper tumor regions limits their effectiveness against tumor cells that are
protected by these adaptive mechanisms. Recent research has focused on
overcoming TME-mediated resistance by developing combination therapies, where
nanoparticles carry not only chemotherapeutic agents but also agents that
modulate the TME, such as immune checkpoint inhibitors or agents that disrupt
the extracellular matrix. Such approaches aim to re-sensitize tumors to
conventional therapies by modifying the tumor's resistance mechanisms and
improving the overall efficacy of nanomedicine(20).
6.4
Limitations of Current Clinical Translation and Regulatory Challenges
Despite the promising results seen in
preclinical models, the clinical translation of nanomedicines has been slow due
to several factors, including regulatory hurdles, manufacturing challenges, and
inconsistent therapeutic outcomes. One of the most significant barriers to
regulatory approval is the lack of standardized guidelines for evaluating the
safety and efficacy of nanomedicines. Regulatory bodies, such as the U.S. Food
and Drug Administration (FDA) and the European Medicines Agency (EMA), have yet
to establish uniform standards for nanomaterial characterization, leading to
variability in clinical trial outcomes. Furthermore, scaling up the production
of nanoparticles for clinical use presents its own set of challenges. The
synthesis of nanoparticles in large quantities while maintaining consistent
size, charge, and functionalization is technically demanding. Additionally,
ensuring that these nanoparticles remain stable and retain their efficacy
during storage and delivery is crucial for successful clinical application.
The potential for long-term effects of nanocarriers, including their
interaction with the immune system and their biodegradation products,
complicates regulatory approval. Safety concerns regarding the unintended
accumulation of nanoparticles in organs or the environment further delay their
widespread clinical use. As such, future research must focus not only on
developing more effective nanomaterials but also on refining the regulatory
frameworks and manufacturing processes to ensure that these treatments are both
safe and scalable for clinical use(22).
7. CASE STUDIES AND CLINICAL TRIALS IN NANOMEDICINE
FOR TARGETING THE TUMOR MICROENVIRONMENT (TME)
Nanomedicine has rapidly advanced in
oncology, offering novel therapeutic strategies aimed at overcoming the
challenges of cancer treatment. Specifically, targeting the tumor
microenvironment (TME) with nanotherapeutics is emerging as a promising approach.
This section reviews successful case studies, ongoing clinical trials, and the
lessons learned from setbacks in the translation of nanomedicine from
preclinical models to clinical practice(8).
7.1
Successful Applications of Nanomedicine in Targeting the Tumor Microenvironment
(TME)
A. Tumor-Targeted
Nanoparticles for Drug Delivery: Nanoparticle-based
drug delivery systems (DDS) have shown significant success in clinical
applications due to their ability to overcome the limitations of traditional
chemotherapy. For example, liposomal formulations like Doxil® (liposomal doxorubicin) have revolutionized the
treatment of metastatic cancers. These systems leverage the Enhanced Permeability and Retention (EPR) effect to selectively accumulate in tumor tissues, resulting in reduced
systemic toxicity. Recent advancements have also focused on the use of polymeric micelles and nanocapsules, which have been
shown to improve the pharmacokinetics and reduce side effects, particularly in
cancers with poorly permeable vasculature and lymphatic drainage(22).
B. Nanoparticles for
Immune Modulation: In addition to drug
delivery, nanomedicine is increasingly being explored for immune modulation
within the TME. Nanoparticles can be engineered to stimulate immune responses,
reprogramming the immune landscape from an immunosuppressive to an immune-activating
environment. Notably, the use of nanoparticles
containing immune adjuvants such as STING agonists has shown
promise in reactivating the immune system and enhancing the effectiveness of immune checkpoint inhibitors
(e.g., PD-1/PD-L1 inhibitors). This approach can potentially transform “cold”
tumors into “hot” tumors, making them more susceptible to immune attack(23).
C. Nanoparticles for
Modulating Tumor Vasculature and Hypoxia: Many
solid tumors are characterized by regions of hypoxia due to aberrant blood vessel formation. Nanoparticles targeting
hypoxic areas of tumors have been developed to release therapeutic agents
specifically in these regions, improving the drug’s effectiveness where it is
most needed. Hypoxia-sensitive
nanoparticles, which release their payload
under low-oxygen conditions, represent an innovative strategy for enhancing the
efficacy of chemotherapy and radiotherapy. These advancements aim to address
the inefficiencies of drug delivery in hypoxic tumor areas, where conventional
therapies often fail(24).
7.2
Ongoing Clinical Trials: Key Findings and Future Directions
Several ongoing clinical trials are
investigating the potential of nanotherapeutics in targeting the TME. These
trials explore various aspects of nanomedicine, from drug delivery to immune
modulation, with promising results and significant insights into future
directions(24).
A.
Nano-Drug Delivery
Systems in Clinical Trials: Nanocarriers such
as albumin-bound paclitaxel (Abraxane) have been extensively tested in clinical trials and have shown
improved outcomes compared to traditional formulations. These
nanoparticle-based drugs offer enhanced drug delivery to tumors while
minimizing systemic toxicity. Abraxane, for example, has been evaluated in
multiple cancers, including breast, lung, and pancreatic cancers, and continues
to show promising efficacy due to its ability to overcome the limitations of
traditional chemotherapy.
B.
Nanoparticles for
Combination with Immunotherapy: The
combination of nanoparticles with
immune checkpoint inhibitors is currently
under investigation in various clinical trials. One such study focuses on the
use of nanoparticle-delivered STING agonists combined with anti-PD-1 therapies in melanoma patients. Early-phase trials have indicated that this
combination may enhance immune activation and improve treatment responses,
particularly in tumors resistant to conventional immune checkpoint therapy.
These findings underscore the potential of nanomedicine in augmenting
immune-based therapies.
C.
Personalized
Nanomedicine Approaches: Future directions in
nanomedicine for cancer treatment are increasingly focusing on personalized or
precision approaches. By using technologies like patient-derived xenografts (PDXs) or organ-on-chip models, researchers can tailor nanotherapeutic strategies to the
individual characteristics of a patient's tumor. These personalized approaches
aim to optimize drug delivery by addressing specific TME features, such as
hypoxia, acidity, and immune suppression. This shift toward precision medicine
is expected to enhance treatment outcomes while minimizing adverse effects(25).
7.3
Lessons Learned from Failures and Setbacks
Despite the significant advancements in
nanomedicine, clinical trials have highlighted several setbacks and challenges
in translating preclinical successes to real-world applications.
A.
Tumor Heterogeneity and
EPR Effect Limitations: The heterogeneity of tumors remains
a significant challenge for nanomedicine. While nanoparticles rely heavily on
the EPR effect for passive targeting, the abnormal vasculature and high
interstitial fluid pressure of many tumors lead to heterogeneous drug distribution.
This variability can result in suboptimal drug delivery, especially in tumors
with poor blood vessel formation. Additionally, the EPR effect is not always
consistent across tumor types, which limits the universal application of this
strategy. Therefore, alternative approaches, such as active targeting using
surface-functionalized nanoparticles or stimuli-responsive
systems, are being developed to improve
targeting precision(15).
B.
Nanotoxicity and Safety
Concerns: Nanoparticles, particularly those
that accumulate in non-target organs such as the liver and spleen, have raised
concerns regarding nanotoxicity. Clinical trials have revealed that while some nanoparticle systems
are biodegradable, others may persist in the body, leading to long-term safety
risks. The immune system's recognition of
nanoparticles as foreign bodies also poses
challenges, necessitating the development of nanoparticles with improved biocompatibility and
more efficient clearance mechanisms. Ensuring the safety of nanotherapeutic
agents through rigorous preclinical testing and careful monitoring during
clinical trials is crucial for advancing these treatments(16).
C.
Immune Suppression in
the TME: Despite the progress in using
nanoparticles to modulate the immune environment, immune suppression remains a
major obstacle. The TME often contains immunosuppressive
cells like regulatory T cells (Tregs) and
myeloid-derived suppressor cells (MDSCs), which inhibit the efficacy of
immune-based therapies. Although nanoparticles can enhance immune responses,
the complex immunosuppressive network within the TME continues to limit the effectiveness of
immunotherapies. Ongoing research is focused on combining immune modulatory nanoparticles
with other treatments to address this issue and shift the TME towards an
immune-activated state(15).
Future
Perspectives
The future of nanomedicine for targeting
the tumor microenvironment (TME) holds great promise with advancements in
multi-functional nanoparticles, artificial intelligence (AI), and machine
learning (ML) for more precise and effective treatments. Emerging technologies,
such as stimuli-responsive and multi-targeting nanoparticles, offer innovative
ways to enhance drug delivery and address multiple TME challenges
simultaneously. AI and ML are increasingly pivotal in optimizing nanoparticle
design and enabling personalized therapies by predicting patient-specific
responses. However, bridging the gap between preclinical research and clinical
application requires overcoming challenges related to scalability, safety, and
regulatory approval. As these hurdles are addressed, the integration of
nanomedicine into clinical practice could revolutionize cancer treatment by
offering more personalized, effective, and less toxic therapies.
CONCLUSION
In
recent years, the application of nanotechnology in cancer therapy has
transformed the landscape of oncology, offering unprecedented opportunities for
improving therapeutic outcomes. By specifically targeting the tumor
microenvironment (TME), nanotherapeutics are paving the way for more precise,
less toxic, and more effective treatments. The TME, with its complex cellular
and molecular components such as immune cells, fibroblasts, and the
extracellular matrix (ECM), presents a formidable barrier to conventional
cancer therapies. Nanomedicine, however, leverages unique properties such as
size, surface functionalization, and responsiveness to specific stimuli to
navigate these challenges. Despite significant progress in the design of
advanced nanocarriers, many challenges remain. The heterogeneity of the TME,
coupled with issues such as poor drug penetration, immune suppression, and drug
resistance, complicates the effectiveness of nano-based therapies. Furthermore,
while clinical trials have demonstrated promising results, the translation of
these innovations from the laboratory to real-world clinical settings is slow
due to regulatory hurdles, manufacturing challenges, and concerns about safety
and toxicity. However, the integration of multidisciplinary
approaches—including the combination of nanotherapeutics with immunotherapies,
the use of stimuli-responsive systems, and advancements in diagnostic and
imaging technologies—holds immense potential to overcome these barriers.
Looking ahead, continued advancements in nanotechnology, coupled with a deeper
understanding of the TME, will likely lead to the development of even more
sophisticated therapies. The future of cancer treatment lies in the ability to
design multifunctional nanomedicines that can not only target tumor cells more
efficiently but also reprogram the TME to enhance therapeutic responses. As
these innovations continue to evolve, there is a growing hope that
nanotherapeutics will become a cornerstone of personalized, precision medicine,
ultimately improving the prognosis and quality of life for cancer patients. The
promising synergy between nanotechnology and cancer therapy provides a powerful
avenue for addressing the complexities of the TME and advancing the fight
against cancer.
REFERENCES
1. Goldman A,
Kulkarni A, Kohandel M, Pandey P, Rao P, Natarajan SK, et al. Rationally
Designed 2-in-1 Nanoparticles Can Overcome Adaptive Resistance in Cancer. ACS
Nano [Internet]. 2016 Jun 28 [cited 2024 Dec 11];10(6):5823–34. Available from:
https://pubmed.ncbi.nlm.nih.gov/27257911/
2. Han S, Chi Y,
Yang Z, Ma J, Wang L. Tumor Microenvironment Regulation and Cancer Targeting
Therapy Based on Nanoparticles. J Funct Biomater. 2023 Feb 28;14(3):136.
3. Adityan S,
Tran M, Bhavsar C, Wu SY. Nano-therapeutics for modulating the tumour
microenvironment: Design, development, and clinical translation. J Control
Release [Internet]. 2020;327(April):512–32. Available from:
https://doi.org/10.1016/j.jconrel.2020.08.016
4. Zheng R, Zhang
S, Zeng H, Wang S, Sun K, Chen R, et al. Cancer incidence and mortality in
China, 2016. J Natl Cancer Cent. 2022 Mar 1;2(1):1–9.
5. Al-Zoubi MS,
Al-Zoubi RM. Nanomedicine tactics in cancer treatment: Challenge and hope. Crit
Rev Oncol Hematol. 2022 Jun 1;174:103677.
6. Raju GSR,
Pavitra E, Varaprasad GL, Bandaru SS, Nagaraju GP, Farran B, et al.
Nanoparticles mediated tumor microenvironment modulation: current advances and
applications. J Nanobiotechnology [Internet]. 2022 Dec 1 [cited 2024 Dec
11];20(1):1–20. Available from:
https://jnanobiotechnology.biomedcentral.com/articles/10.1186/s12951-022-01476-9
7. Wu T, Dai Y.
Tumor microenvironment and therapeutic response. Cancer Lett [Internet]. 2017
Feb 28 [cited 2024 Dec 11];387:61–8. Available from:
https://pubmed.ncbi.nlm.nih.gov/26845449/
8. Feng Y, Tang
Q, Wang B, Yang Q, Zhang Y, Lei L, et al. Targeting the tumor microenvironment
with biomaterials for enhanced immunotherapeutic efficacy. J Nanobiotechnology
[Internet]. 2024 Dec 1 [cited 2024 Dec 11];22(1). Available from:
https://pubmed.ncbi.nlm.nih.gov/39605063/
9. Kim J, Hong J,
Lee J, Fakhraei Lahiji S, Kim YH. Recent advances in tumor
microenvironment-targeted nanomedicine delivery approaches to overcome
limitations of immune checkpoint blockade-based immunotherapy. J Control
Release. 2021 Apr 10;332:109–26.
10. Shen S, Gao Y,
Ouyang Z, Jia B, Shen M, Shi X. Photothermal-triggered dendrimer nanovaccines
boost systemic antitumor immunity. J Control Release. 2023 Mar 1;355:171–83.
11. Afzal M,
Ameeduzzafar, Alharbi KS, Alruwaili NK, Al-Abassi FA, Al-Malki AAL, et al.
Nanomedicine in treatment of breast cancer – A challenge to conventional
therapy. Semin Cancer Biol. 2021 Feb 1;69:279–92.
12. Bussard KM,
Mutkus L, Stumpf K, Gomez-Manzano C, Marini FC. Tumor-associated stromal cells
as key contributors to the tumor microenvironment. Breast Cancer Res
[Internet]. 2016 Aug 11 [cited 2024 Dec 11];18(1). Available from:
https://pubmed.ncbi.nlm.nih.gov/27515302/
13. Jin C, Wang K,
Oppong-Gyebi A, Hu J. Application of nanotechnology in cancer diagnosis and
therapy - A mini-review. Int J Med Sci. 2020;17(18):2964–73.
14. Nguyen VH, Kim
HS, Ha JM, Hong Y, Choy HE, Min JJ. Genetically engineered Salmonella
typhimurium as an imageable therapeutic probe for cancer. Cancer Res
[Internet]. 2010 Jan 1 [cited 2024 Dec 11];70(1):18–23. Available from:
https://pubmed.ncbi.nlm.nih.gov/20028866/
15. Mosleh-Shirazi
S, Abbasi M, Moaddeli MR, Vaez A, Shafiee M, Kasaee SR, et al. Nanotechnology
Advances in the Detection and Treatment of Cancer: An Overview.
Nanotheranostics. 2022;6(4):400–23.
16. Dai Q,
Bertleff-Zieschang N, Braunger JA, Björnmalm M, Cortez-Jugo C, Caruso F.
Particle Targeting in Complex Biological Media. Adv Healthc Mater. 2018 Jan
10;7(1).
17. Navigating the
Future: Advancements in Monoclonal Antibody Nanoparticle Therapy for Cancer. J
Drug Deliv Sci Technol. 2024 Dec 6;106495.
18. Ray P. Curing
Cancer with Nanotherapy Continues to be an Elusive Goal. J Immunol Sci.
2021;5(2):36–9.
19. Zeng L, Gowda
BHJ, Ahmed MG, Abourehab MAS, Chen ZS, Zhang C, et al. Advancements in
nanoparticle-based treatment approaches for skin cancer therapy. Mol Cancer
[Internet]. 2023 Dec 1 [cited 2024 Dec 11];22(1). Available from:
https://pubmed.ncbi.nlm.nih.gov/36635761/
20. Chen L, Hong W,
Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based
on biomimetic nanoparticles. Signal Transduct Target Ther [Internet]. 2021 Dec
1 [cited 2024 Dec 11];6(1). Available from:
https://pubmed.ncbi.nlm.nih.gov/34099630/
21. Siegel RL,
Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin
[Internet]. 2022 Jan [cited 2024 Dec 11];72(1):7–33. Available from:
https://pubmed.ncbi.nlm.nih.gov/35020204/
22. Kulkarni JA,
Witzigmann D, Chen S, Cullis PR, Van Der Meel R. Lipid Nanoparticle Technology
for Clinical Translation of siRNA Therapeutics. Acc Chem Res. 2019 Sep
17;52(9):2435–44.
23. Riley RS, June
CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat
Rev Drug Discov 2018 183 [Internet]. 2019 Jan 8 [cited 2024 Dec
11];18(3):175–96. Available from:
https://www.nature.com/articles/s41573-018-0006-z
24. Lakshmanan VK,
Jindal S, Packirisamy G, Ojha S, Lian S, Kaushik A, et al. Nanomedicine-based
cancer immunotherapy: recent trends and future perspectives. Cancer Gene Ther.
2021 Sep 1;28(9):911–23.
25. Karahan HE,
Wiraja C, Xu C, Wei J, Wang Y, Wang L, et al. Graphene Materials in
Antimicrobial Nanomedicine: Current Status and Future Perspectives. Adv Healthc
Mater. 2018 Jul 11;7(13).