In the fight against cancer, half the battle is won on the basis of its early detection. Nanotechnology offers new molecular contrast agents and materials to enable earlier and more accurate initial diagnosis as well as continuous monitoring of cancer patients' treatment.
While nanoparticles are not yet used clinically for cancer detection or diagnosis, they are already on the market in various medical screenings and tests, with the most widespread use of gold nanoparticles being in home pregnancy tests. Nanoparticles are also at the heart of Nanosphere's Verigene® system and T2 Biosystems' T2MR system, which are currently used in hospitals for a variety of indications.
In cancer, nanodevices are being studied to detect blood-borne biomarkers, including cancer-associated proteins circulating in tumor cells, circulating tumor DNA, and exosomes shed by tumors. Nano-enabled sensors are capable of highly sensitive, specific and multiplexed measurements. State-of-the-art equipment combines detection with genetic analysis to further elucidate a patient's cancer and possible treatments and disease progression.
Nanoparticles, already clinically established as anatomical structure contrast agents, are being developed to act as molecular imaging agents, displaying the presence of genetic mutations relevant to cancer or the functional properties of tumor cells. This information may be used to select a course of treatment or change a treatment plan. Bioactivatable nanoparticles, which change their properties in response to factors or processes in the body, act as dynamic reporters of in vivo states and can provide spatial and temporal information about disease progression and therapeutic response.
BildLive
Current imaging techniques can only detect cancer after causing a visible change in a tissue, by which time thousands of cells have proliferated and perhaps metastasized. And even if it is visible, the type of tumor - malignant or benign - and the characteristics that might make it responsive to a particular treatment need to be evaluated through tissue biopsies. Although some primary malignancies can be determined as metastatic, tumors arising from metastatic sites and micrometastases are extremely difficult to detect with modern imaging techniques, despite the known tissues in which they commonly occur.First. Finally, surgical resection of tumor tissue remains the standard of care for many types of tumors, and surgeons must weigh the consequences of removing healthy, often vital, tissue versus the unevenly grown cancerous mass. Ultimately, with current surgical techniques, it is not possible to remove cancer cells at the single cell level.
Nanotechnology-based imaging contrast agents being developed and implemented today offer the opportunity to target and significantly improve in vivo tumor detection by traditional scanning devices such as magnetic resonance imaging (MRI) (PET) and computed tomography (CT). ). In addition, current nanoscale imaging platforms enable new non-traditional imaging modalities used for clinical cancer treatment and diagnosis, e.g. B. photoacoustic tomography (PAT), Raman spectroscopic imaging and multimodal imaging (i.e. specific contrast agents for several imaging modalities simultaneously). ). Nanotechnology enables all of these platforms through their ability to deliver multiple components simultaneously (e.g., cancer cell-specific targeting agents or traditional imaging contrast agents) and nanoscale materials, which are the very contrast agents that enable signal.
NCI-funded research has produced many notable examples in recent years. For example, researchers at Stanford University and Memorial Sloan Kettering Cancer Center have developed multimodal nanoparticles capable of delineating the edges of brain tumors preoperatively and intraoperatively. These MRI-PAT Raman nanoparticles can be used for both tumor growth tracking and surgical staging by MRI, but also in the same particle used during surgical resection of the brain tumor to give the surgeon "eyes" down to the individual at the cancer cell level, increasing the potential for tumor-specific tissue removal.
For metastatic melanoma, researchers at MSKCC and Cornell University have developed hybrid silica nanoparticles ('C-dots') that offer optical contrast and PET image contrast on the same platform. These nanoparticles actively target cancer with cRGDY peptides that target this specific tumor type and have done so successfully in early clinical trials.
Another clinical cancer imaging problem addressed by nanoscale solutions is prostate cancer. Stanford University researchers have recently developed nanotechnologies that provide the anatomical size and location of prostate cancer cells (nanobubbles for ultrasound imaging) and functional information to prevent overdiagnosis/treatment and monitor progress (self-assembling nanoparticles for photoacoustic imaging). The nanoplatforms developed by this group are directly coupled to their recently approved handheld transrectal photoacoustic ultrasound (TUSPA) device. Ultimately, it offers a more effective, integrated and less invasive prostate cancer imaging and biopsy technique for diagnosis and prognosis prior to performing common procedures (surgical resection, radiation therapy, etc.).
Similarly, gold nanoparticles are used to improve light scattering for endoscopic techniques that can be used during colonoscopy. A really powerful potential that has always been envisioned for nanotechnology in cancer has been the potential to simultaneously image and deliver therapy in vivo, and various groups have been pushing these nanoscale “theranostic” platforms. A group at Emory University developed one for ovarian and pancreatic cancers, which are traditionally more difficult to treat. Its pancreatic cancer platform can destroy the fibrotic stromal tissue that protects these tumors in the pancreas from. After crossing this barrier, they consist of magnetic iron cores that allow MRI contrast for diagnosis and deliver small molecule drugs directly to cancer cells for treatment.
Finally, nanotechnology enables the visualization of molecular markers that identify specific stages and therapy-induced cancer cell death, allowing physicians to see cells and molecules undetectable by traditional imaging. A group at Stanford developed the Target-Enabled in Situ Ligand Assembly (TESLA) nanoparticle system. This is based on nanoparticles that form directly in the body after intravenous injection of molecular precursors. Precursors contain specific sequences of atoms that can only form larger nanoparticles after being cleaved by enzymes produced by cancer cells during apoptosis (i.e. cell death) and carry various imaging contrast agents to monitor the local tumor response (PET , MRI etc.). to therapies. The ability to follow cancer cell death in vivo and at the molecular level is extremely important to provide effective dosing regimens and/or to accurately administer new therapies or combinations.
In vitro detection
activated for nanotechnologyIn-vitro-Diagnosticsoffer high sensitivity and selectivity, as well as the ability to perform simultaneous measurements on multiple targets. Well-established manufacturing techniques (e.g., lithography) can be used to create integrated handheld devices or point-of-care systems. A diagnostic device or biosensor contains a biological recognition element that can detect the presence, activity, or concentration of a specific biological molecule in solution through a biochemical reaction. This reaction may involve, for example: antigen and antibody binding, hybridization of two single-stranded DNA fragments, or capture ligand binding to the cell surface epitope. A transducer portion of the sensor device is used to convert the biochemical event into a quantifiable signal that can be measured. Transmission mechanisms can be based on light, magnetic or electronic effects.
Various devices have been developed for the detection of various serum or tissue biological signatures. Some examples of nanotechnology or nanoparticle based diagnostic devices are shown in Figure #. The barcode bioassay was developed as a sandwich immunoassay in the laboratory of Chad Mirkin at Northwestern University. It uses magnetic nanoparticles (MMPs) functionalized with monoclonal antibodies specific to the target protein of interest, which are then mixed with the sample to promote target protein capture. The hybrid MMP-protein structures are then combined with gold nanoparticle (Au-NP) probes bearing DNA barcodes. Target protein-specific DNA barcodes are released into solution and detected using the scanometric assay with sensitivities in the femtopicomolar range.
James Heath's lab at Caltech has developed sandwich immunoassay devices based on DNA-encoded antibody libraries (DEAL). The DEAL technique uses DNA-directed immobilization of antibodies in microfluidic channels, allowing a pre-patterned single-stranded (ss) DNA barcode microarray to be converted into an antibody microarray. The ssDNA oligomers attached to the sensor surface are robust and can withstand high channel fabrication temperatures. Subsequent flow of the DNA-antibody conjugates into the channels converts the DNA microarray into an antibody microarray and allows the performance of surface-binding multiplex sandwich immunoassays. These devices enable on-chip blood separation and measurement of large amounts of protein directly from blood.
The diagnostic magnetic resonance imaging (DMR) sensor platform was developed in Ralph Weissleder's laboratory at Massachusetts General Hospital. The DMR mechanism uses changes in the transverse relaxation signal of water molecules in a magnetic field as a detection mechanism for analytes labeled with magnetic nanoparticles. Highly integrated systems, including microfluidic processing circuits and high signal-to-noise ratio nuclear magnetic resonance (NMR) acquisition heads, have been built and are capable of detecting the presence of cells, vesicles and proteins in clinical samples.
Shan Wang's lab at Stanford University has developed giant magnetoresistive (GMR) biosensors for protein detection. These nanosensors work by changing their electrical resistance in response to changes in the local magnetic field. They were adapted to detect biological signatures in solution and implemented a traditional sandwich assay directly on GMR nanosensors. Antibodies are immobilized on the surface of the GMR sensor and serve as capture probes for the sample containing target proteins. A magnetic particle is used to tag the biomolecule of interest in the sample and the GMR sensor is used for signal transmission. These sensors have been used to measure protein concentrations in complex sample mixtures and have also been used to assess the kinetics of protein interactions.
The devices described above are capable of analyzing large panels of biological signatures while providing a high level of multiplexing. Data analysis can establish correlations between different levels of biomarkers and map signal correlations of networks, thus providing tools for patient stratification based on their response to different treatments and ultimately improving the therapeutic efficacy of the selected treatment. Recent advances in microfluidic technologies have opened up opportunities to integrate sample preparation and processing with biosensors, creating fully integrated devices that provide complete data from a single sample directly for medical diagnosis.
Measurement of response to therapy and liquid biopsy
Measuring an individual patient's response to therapy over the course of their illness is the basis for accurate medical care and prognosis. Accurate and relevant disease monitoring can enable optimized treatment regimens (e.g., correction of therapeutic course, drug combinations, and dose reduction), preventive clinical decision-making (e.g., therapeutic responders versus non-responders and more), and patient-to-clinic stratification of trials. In addition to the more traditional gold standards of in vivo imaging, tissue biopsy, and in vitro diagnostics available for this purpose, "liquid biopsy" offers the ability to measure response to therapy through single and serial blood draws. Conventional biopsies involve the direct resection of small tumor tissue volumes and therefore remain invasive procedures that cannot provide the sample collection necessary to follow the course of the disease in relation to the course of therapy or the dynamics of its evolving biology. Liquid biopsies rely on the fact that tumors release material (eg, cells, DNA, other cancer-specific biomolecules) into the circulation over time and in response to therapy. Although the amount of materials eliminated from a given tumor and/or stage is typically at incredibly low concentrations compared to the rest of the blood components (e.g. erythrocytes, leukocytes, platelets, plasma, etc.). This requires specific and sensitive tools to detect, capture and purify circulating tumor material from the rest. Nanotechnology is making it possible for these tools to become a reality.
Recent technological advances in coupling complex microfluidics and nanoscale materials have enabled high-purity detection and downstream functional characterization of circulating tumor cells (CTCs), cell-free tumor DNA, microemboli, exosomes, proteins, neoantigens, and more. Recent examples include the capture and subsequent release of CTCs in microfluidic systems to keep cells viable for downstream whole genome sequencing, ex vivo expansion, RNA sequencing, and more. Of these examples, one type of device uses magnetic nanoparticles to enrich whole blood prior to magnetic separation within the microfluidic, and the other device uses thermoresponsive nanopolymers that specifically capture CTCs as they flow through the microfluidic, and then are released once again after a temperature change blood is present processing is complete. In both cases the detection sensitivities are very high (e.g. for enumeration > 95%) and the detection purity is much higher than other non-nanomaterials based devices. In addition, processing times are increasing each year as technology advances, currently averaging 10 mL of blood per 30 minutes.
FAQs
How is nanotechnology used in cancer detection? ›
Nanotechnology can provide rapid and sensitive detection of cancer-related molecules, enabling scientists to detect molecular changes even when they occur only in a small percentage of cells. Nanotechnology also has the potential to generate entirely novel and highly effective therapeutic agents.
What is nanosensors for early cancer detection? ›Nanosensors will enable the screening of normal/diseased individuals among the population by the early detection of biomarkers, circulating tumor cells or exosomes from blood samples.
Can nanotechnology detect early diseases? ›Nanoparticles can attach to proteins or other molecules, allowing detection of disease indicators in a lab sample at a very early stage.
What are early detection methods for cancer? ›Imaging tests used in diagnosing cancer may include a computerized tomography (CT) scan, bone scan, magnetic resonance imaging (MRI), positron emission tomography (PET) scan, ultrasound and X-ray, among others. Biopsy. During a biopsy, your doctor collects a sample of cells for testing in the laboratory.
What are the disadvantages of nanotechnology in cancer diagnosis? ›However, this technique has disadvantages, such as its lethal effect on normal cells within the body. Accordingly, therapy provided for the tumor cells is lethal for the normal cells, leading to neural toxicity, suppression of bone marrow and cardiomyopathy, etc.
What are 3 uses of nanotechnology? ›- Electronics.
- Energy.
- Biomedicine.
- Environment.
- Food.
- Textile.
As far back as 1995, nanotechnology has offered clinicians novel tools to treat patients. Find a list of the currently approved nano-enabled therapeutics here.
Why is it better to detect cancer early? ›When cancer care is delayed or inaccessible there is a lower chance of survival, greater problems associated with treatment and higher costs of care. Early diagnosis improves cancer outcomes by providing care at the earliest possible stage and is therefore an important public health strategy in all settings.
What is nanoparticles to cure cancer? ›Nanoparticles (1–100 nm) can be used to treat cancer due to their specific advantages such as biocompatibility, reduced toxicity, more excellent stability, enhanced permeability and retention effect, and precise targeting. Nanoparticles are classified into several main categories.
What is the future of early cancer detection? ›In the future, advances in sensors, contrast agents, molecular methods, and artificial intelligence will help detect cancer-specific signals in real time. To reduce the burden of cancer on society, risk-based detection and prevention needs to be cost effective and widely accessible.
What are 5 ways to detect cancer? ›
- CT scan. A CT scan uses an x-ray machine linked to a computer to take a series of pictures of your organs from different angles. ...
- MRI. An MRI uses a powerful magnet and radio waves to take pictures of your body in slices. ...
- Nuclear scan. ...
- Bone scan. ...
- PET scan. ...
- Ultrasound.
Cancer is nearly always diagnosed by an expert who has looked at cell or tissue samples under a microscope. In some cases, tests done on the cells' proteins, DNA, and RNA can help tell doctors if there's cancer. These test results are very important when choosing the best treatment options.
What are human risks of nanotechnology? ›Nanoparticles can get into the body through the skin, lungs and digestive system. This may help create 'free radicals' which can cause cell damage and damage to the DNA. There is also concern that once nanoparticles are in the bloodstream they will be able to cross the blood-brain barrier.
Which technique is used for early diagnosis of disease? ›Reason: Recombinant DNA technology, PCR and ELISA are some of the techniques that serve the purpose of early diagnosis.
How is early detection of disease done? ›A screening test is done to detect potential health disorders or diseases in people who do not have any symptoms of disease. The goal is early detection and lifestyle changes or surveillance, to reduce the risk of disease, or to detect it early enough to treat it most effectively.
What are the four 4 benefits and concerns of using nanotechnology? ›Nanotechnology offers the potential for new and faster kinds of computers, more efficient power sources and life-saving medical treatments. Potential disadvantages include economic disruption and possible threats to security, privacy, health and the environment.
What are the risks of nanotechnology in cancer treatment? ›Current nanotechnology-based treatments such as Abraxane and Doxil do cause side effects like weight loss, nausea, and diarrhea. But these problems may be from the chemotherapy drugs they contain. Researchers should learn more about the side effects of these treatments as they study them in clinical trials.
Does nanotechnology has a positive or negative impacts? ›Nanotechnology can cause positive and significant changes to air quality, water quality, and sustainable energy generation. It can help us to repair the environment and save it.
What is an example of nanotechnology being used today? ›Nano-engineered materials make superior household products such as degreasers and stain removers; environmental sensors, air purifiers, and filters; antibacterial cleansers; and specialized paints and sealing products, such a self-cleaning house paints that resist dirt and marks.
What is the most used nanotechnology? ›Silver is the most common nano-material used in products, followed by carbon-based nano-materials and metal oxides such as TiO2. Nanotechnology is going to pave the way for a revolution in materials, information and communication technology, medicine, genetics, etc.
How do nanobots detect cancer cells? ›
The nanorobot is designed with blood energy harvesting capability and the accumulation of electricity in a capacitor, which forms the main body of the nanorobot. Glucose hunger-based cancer detectors immobilized on a carbon nanotube sensor, reduces its electrical resistance when attached to a cancer cell.
How do scientists use nanobots to treat cancer? ›Unlike chemotherapy and radiation therapy, which destroys healthy cells in addition to cancer cells, nanobots release cancer-fighting drugs directly into the malignant tumor. In this way, nanobots maximize the efficiency of the drugs while greatly reducing harm to surrounding healthy tissue.
What is the impact factor of cancer nanotechnology 2022? ›Cancer Nanotechnology Impact Factor 2022
The latest impact factor of Cancer Nanotechnology is 7.917. The impact factor (IF) is a measure of the frequency with which the average article in a journal has been cited in a particular year.
Cancer that's diagnosed at an early stage, when it isn't too large and hasn't spread, is more likely to be treated successfully.
What is the earliest diagnosis of cancer? ›Our oldest description of cancer (although the word cancer was not used) was discovered in Egypt and dates back to about 3000 BC. It's called the Edwin Smith Papyrus and is a copy of part of an ancient Egyptian textbook on trauma surgery.
Who cured cancer with nanoparticles? ›Out of three human studies, only one showed a passage of inhaled nanoparticles into the bloodstream. Materials which by themselves are not very harmful could be toxic if they are inhaled in the form of nanoparticles. The effects of inhaled nanoparticles in the body may include lung inflammation and heart problems.
How long do nanoparticles stay in the body? ›Unlike conventional imaging agents and therapeutics, many nanoparticles are highly stable in vivo—exemplified by a recent study suggested that quantum dots may be retained in the body (and remain fluorescent) for more than 100 days [2].
How are nanoshells used in cancer detection? ›The nanoshell can be stimulated with infrared light in which the nanoshell absorbs the energy of the light and consequently generates heat. Through this mechanism, nanoshells have a potential photothermal therapeutic ability to destroy bacteria and tumor cells.
When was nanotechnology used in cancer treatment? ›A variety of targeting agents such as monoclonal antibodies (mAbs) and nucleic acids (aptamers) are also used to enhance tumoral uptake of nanoparticles. Using mAbs for targeting in cancer therapy was first described by Milstein in 1981 (Warenius et al., 1981).
What is a way nanobots are being used to treat cancer? ›
A nanorobot can aid with smart chemotherapy for medication administration and give an efficient early dissolution of cancer by targeting only the neoplastic-specific cells and tissues and preventing the surrounding healthy cells from the toxicity of the chemotherapy drugs so being used.
What device can detect cancer? ›During the CT scan, you will lie still on a table that slides into a donut-shaped scanner. The CT machine moves around you, taking pictures. Learn more about CT scans and how they are used to diagnose cancer.
Which nanoparticles are best for cancer? ›Silica Nanoparticles
Mesoporous silica NPs are considered one of the best drug carriers due to their better pharmacokinetic properties. They have been extensively used in immunotherapy. According to a study, colorectal cancer cells have shown successful uptake of camptothecin-loaded mesoporous silica NPs.
Carbon nanotubes have become popular tools due to their unique physicochemical properties in cancer diagnosis. These are considered as the most promising nanomaterials with the capability of both detecting the cancerous cells and delivering drugs or small therapeutic molecules to cancer cells.
What can nanosensors detect? ›Nanosensors are able to detect the presence of gases, aromas, chemical contaminants, pathogens, and even changes in environmental conditions.
Can nanobots detect cancer? ›The applications of nanobots can even extend as far as cancer diagnosis and treatment. According to the IFL Science article, scientists have been testing nanobots to seek out and destroy cancer cells actively.
Are nano robots designed to destroy cancer cells? ›Nanomedicine researchers have successfully programmed nanorobots to find tumors and cut off their blood supply while leaving healthy tissue unharmed. They're microscopic, autonomous, and on a mission. They are nanorobots programmed to seek and destroy tumors.
Are nanobots used in medicine today? ›Nanorobots are being used in various domains of pre-clinical and clinical medicine. In pre-clinical medicine, nanorobots are being employed in bioimaging and various delivery systems of drugs, gene therapy, living cells, and inorganic therapeutics.