According to Statistics Canada, gastrointestinal (GI) diseases are responsible for over 300,000 deaths each year in Canada across all ages (3). GI diseases are predicted to increase over the next ten years and can impact human health on a global level. Common causes of digestive issues within our population involve chronic stress, harmful pesticides, and the consumption of the Standard American Diet (3)(4).
After identifying this increase in disease prevalence, researchers are aware that there is a gap in diagnostic technology assessing GI symptoms. By turning to the world of smart medical devices, we open up a variety of options for GI disease diagnosis. One of these examples is the SmartPill device.
The SmartPill is a wireless capsule that a physician can use which monitors parameters such as pH, pressure, gastrointestinal transit time, and temperature throughout your digestive tract (5). The capsule is currently approved by the FDA for the diagnosis of conditions that are related to gastric emptying delays and general gastrointestinal motility disease (6).
SOURCE: BASS Medical Group (1)
The SmartPill functions as an endoscopic capsule. Patients swallow an activated wireless pH, pressure, and temperature capsule. This capsule contains sensors that measure pH (with a range of 0.5-9), temperature (with a range of 25-49 °C), and pressure (with a range of 0-350 mmHg) (5). After ingestion, the capsule signals are transmitted from within the GI tract and captured by a receiver. The data receiver is a portable device worn on a belt or a lanyard by the patient and it records information collected by the capsule (5). The data is then stored in the device and transmitted to a computer which provides the physician with the necessary information to evaluate the function of the patient’s stomach and intestines. The patient then continues with their day-to-day activities, and the pill is usually passed within 1-2 days. After passing the pill, the patient returns the data recorder to the physician’s office where the results are then analyzed.
SOURCE: Wang et. al (2)
The current standard of care for diagnostic procedures involves invasive or uncomfortable methods such as upper GI barium swallow tests, gastroscopy, endoscopy, or gastric manometry. The use of the SmartPill, however, may mitigate patient discomfort due to its ingestible approach. For the reliability of results, a study was done assessing the clinical use of wireless motility capsules. The SmartPill capsule detected a generalized motility disorder in 51% of patients (7). The capsule was also shown to influence management decisions in 30% of patients with lower GI disorders and 88% of patients with upper GI disorders (8). These results show that the SmartPill can function as an advantage to our healthcare system by comfortably assessing gastrointestinal parameters, allowing seamless data collection, and providing physicians with assistance for disease management.
Browning KN, Travagli RA. Central Nervous System Control of Gastrointestinal Motility and Secretion and Modulation of Gastrointestinal Functions. Compr Physiol. 2014 Oct;4(4):1339–68.
Tu P, Chi L, Bodnar W, Zhang Z, Gao B, Bian X, et al. Gut Microbiome Toxicity: Connecting the Environment and Gut Microbiome-Associated Diseases. Toxics. 2020 Mar 12;8(1):19.
Cassilly D, Kantor S, Knight LC, Maurer AH, Fisher RS, Semler J, et al. Gastric emptying of a non-digestible solid: assessment with simultaneous SmartPill pH and pressure capsule, antroduodenal manometry, gastric emptying scintigraphy. Neurogastroenterol Motil. 2008;20(4):311–9.
Farbod Azaripour Masooleh—McMaster Life Sciences 2025
Gene Editing has been a frequently discussed topic with its increasing importance, as new technologies continue to improve it. CRISPR is one such gene editing technique that has revolutionized the field by making it more precise and easier. This has made scientists hopeful about the possibility of correcting disease-causing genes to prevent genetic disorders. The VI CRISPR-Cas effector, Cas13b, targets designated RNAs directly. The combination of Cas13b and ADAR2 adenosine deaminase domain with rational protein engineering has resulted in a more efficient enzyme. This has made efficient and specific RNA depletion of mammalian cells possible. This system is known as RNA Editing for Programmable A to I Replacement (REPAIR). This system can edit full-length transcripts carrying pathogenic mutations, not limited to specific sequences. To minimize the system and facilitate viral delivery, REPAIR is being modified to increase its specificity. This gives scientists a reliable RNA-editing platform with broad applicability for research studies, therapeutics, and biotechnology advancements. To test this system in humans, REPAIRv1 was developed to correct disease-causing G→A mutations in nucleotides.
Figure 1: measuring the flexibility of the sequences for editing RNA using REPAIRv1. SOURCE: Science
Substantial editing was achieved at 33 sites with an efficiency rate of 28%. The REPAIR system enables multiplex editing of multiple disease-causing variants. The dCas13b platform, designed for programmable RNA binding, can also be used for live transcript imaging, splicing modification, targeted localization of transcripts, RNA-binding protein pulldown, and epitranscriptomic modifications.
Figure 2: This image shows the use and effect of REPAIRv1 on repairing a G→A mutation. SOURCE: Science
The base conversions that scientists are able to achieve by using REPAIR are restricted to using adenosine in order to create inosine. But, combining dCas13 with other RNA editing domains, can help with editing cytidine to uridine. In addition, for relaxing the substrate preference so that cytidine can be targeted, mutagenesis of ADAR can be used. This grants more specificity from the duplexed RNA substrate requirement so that it can aid C to U editors.
In conclusion, as technology advances, the equipment used for scientific purposes gets more precise and accurate. CRISPR, as one such tool, holds great promise for curing genetic diseases and improving millions of lives in the near future.
References
Cox DB, Gootenberg JS, Abudayyeh OO, Franklin B, Kellner MJ, Joung J, et al. RNA editing …..with CRISPR-CAS13. Science. 2017Nov24;358(6366):1019–27.
You may have come across the term designer babies in recent years as the topic has started to rise in popularity. Although they may sound like babies who cruise around town in their Gucci baby stroller, snuggled up in their Louis Vuitton onesie, the reality is much more fascinating. Unfortunately, however, many still need to be made aware of the actual science, methods, and implications of the development of designer babies and how we may already be seeing the application of this technology all around us.
The idea of designer babies was initially proposed to reduce or avoid heritable diseases coded by mutations in our DNA, creating healthy and happier babies (1). This process is done by editing the genome through methods such as preimplantation genetic diagnosis or genetic modification (1).
SOURCE: inviTRA
Preimplantation genetic diagnosis (PGD) is the process of genetic testing of an embryo before it is implanted in the uterus and is used in conjunction with in vitro fertilization (IVF) (2). PGD identifies potential genetic abnormalities in embryos before embryo transfer (implanting a newly formed embryo into the woman’s uterus), commonly used to prevent single-gene conditions such as cystic fibrosis or sickle cell anemia (3). For example, in sickle cell anemia, a single allele point mutation can be targeted. However, more complex conditions such as autism involve the interaction of many genes and the environment and cannot be targeted or even understood as thoroughly. Despite this, PGD is available for all inherited conditions where the exact mutation is known and can be tested on the embryo (3).
The possibilities with PGD are still in development, and although the technique still suffers from limitations, it is a practical and more sensible approach to reducing heritable diseases.
However, the idea of the designer baby in popular culture has taken a new meaning and often refers to the use of genomic editing technologies for purposes beyond simply reducing heritable disease (1). Unlike PGD, the topic of gene editing in humans is an ever-changing debate on the ethics, morality, and beliefs regarding this technology’s political and social implications. Nevertheless, individuals have already begun creating babies with edited genomes and engineered mutations (4).
Individuals may take advantage of this technology to create babies with certain facial features and physical characteristics such as coloured eyes, athletic builds, and taller bodies. However, using genomic editing to create babies with “ideal” features strongly reinforces stereotypes and prejudice against people with features that are not considered a high priority for gene editing. Thus, the use of this technology poses various questions regarding its safety, ethics, and social implications. Furthermore, as this technology is in the early stages of development, it is likely that access to this resource will come at a high cost and will not benefit individuals who lack the necessary finances to support such procedures, widening the gap between the socioeconomic classes seen throughout society. A highly controversial use of genome editing is demonstrated when human genomes are altered using technologies such as CRISPR-Cas9 (6). CRISPR-Cas9 involves genome editing that may be used to make changes to genes in egg or sperm cells or to the genes of an embryo that could be passed to future generations. This application of genome editing raises the previously mentioned ethical concern of passing down enhanced human traits such as height or intelligence and the possible issues with the safety of such technique. In addition, it is possible that the use of CRISPR-Cas9 may produce “off-target” effects and can damage the genome in unpredictable ways.
SOURCE: The Scientist
Currently, genomic editing is conducted using genome editing tools to alter or destroy mutated mitochondrial DNA (1). In the embryo, thousands of copies of mitochondrial DNA (mtDNA) are present in the cell’s cytoplasm, which is highly prone to mutations (1). A high enough concentration of mutated mtDNA will lead to the development of mitochondrial diseases, which genomic editing hopes to reduce. This application of genome editing is widely accepted throughout the scientific community; however, it still comes with risks.
Thus, the creation of designer babies remains in the early stages of development. Although there have been many significant advances in genetic editing and modification in recent decades, the topic still causes many to question if it should even exist and the ethical controversies it proposes (5).
References
Pang T.K. Ronald, Ho, P.C. Pang. Designer Babies [Internet]. 2016;26(2): 59-60. Obstetrics, Gynaecology & Reproductive Medicine. Available from: https://doi.org/10.1016/j.ogrm.2015.11.011
Laura Weiler—Integrated Biomedical Engineering & Health Sciences 2026
Today’s society centers itself around immediacy and efficiency, and these values are also reflected in the world of medical research. Current diagnostic tools are continuously improving thanks to advancements in data manipulation, computer models, and assays.1
Among these advancements are Quenchbodies: fluorescent immunosensors that show promise as a novel, rapid antigen detector for disease diagnosis, food control, and environment monitoring.2
A Quenchbody (Q-body) uses an antibody fragment to detect a specific molecule (called an antigen or analyte) in an immunoassay.2 Antibodies are molecules involved in the immune response that detect a particular analyte, for example, a specific hormone, virus, or vitamin.3
Enzyme-linked immunosorbent assays (ELISAs) are frequently used immunoassays due to their high specificity and accuracy in detecting an analyte.4 A downside to an ELISA is that it requires additional steps to wash away any excess sample or antibodies throughout the process.5
Another type of immunoassay is a fluoroimmunoassay, which labels the antibodies with fluorescent tags and quantifies the antigen concentration using fluorescence (i.e., such that higher fluorescence means higher antigen presence).3 Q-bodies provide an innovative take on fluoroimmunassays due to their simplicity and accuracy.6
What is notable about a Q-body is that it does not involve any additional reagent or separation steps, as is the case with an ELISA.7 Ueda et al.7 discovered a “single chain variable region fragment” (scFv) of a fluorescent-labelled antibody which increased in fluorescence when the label was displaced by an antigen. Ueda et al.7 called it a Q-body: an scFv (also called a fragment antibody (Fab)) and a specific fluorescent label so that presence of the antibody’s antigen can replace the label.7 The name “Quenchbody” comes from the mechanism behind this enhanced fluorescence, which involves a naturally-occurring amino acid called tryptophan (Trp).7 Trp contained in an antibody interacts with the dye in the fluorescent tag and “quenches” or suppresses its fluorescent properties unless an antigen replaces the fluorescent label at the Fab, causing the label to fluoresce, as depicted in Figure 1.8 Generally, a Q-body can be made from any antibody and specific scFvs are readily available, making Q-bodies valuable tools for a variety of applications.7
Figure 1: Quenchbody Mechanism.
SOURCE: Creative Biolabs
The Q-body immunoassay has already been tested for its applications to monitor neonicotinoids for environmental and food safety.9 Neonicotinoids are known for their uses in insecticides, particularly Imidacloprid (ICP), the most popular insecticide worldwide.9 Neonicotinoids lead to paralysis and death of insects at even low doses and contribute to honey bee colony collapse.9 Since crop seeds are often coated with the insecticide before being planted, plants, nectar, and pollen may contain low concentrations of neonicotinoid.9 Conventional methods (gas or liquid chromatography and mass spectrometry) used to quantify pesticide residues are often time-consuming and lack portability.9 This is where using a Quenchbody assay comes in handy. The assay was completed within two minutes and successfully quantified ICP residues with a slightly lower sensitivity than when using an ELISA.9
As for other applications, researchers have been looking at using Q-body to monitor drug misuse, for cancer diagnostics, in vitro or in vivo imaging, and for SARS-CoV2 detection.6,8 Ueda et al.10 concluded that the Quenchbody measured the SARS-CoV2 nucleocapsid protein better than current lateral flow antigen tests. While the functionality and production of the Q-body assay still need to be fine-tuned, it offers a sensitive, simplified, rapid antigen detection system with a promising range of applications in medicine and health.
Ueda H, Dong J. Recent advances in quenchbody, a fluorescent immunosensor. Sensors (Basel) [Internet]. 2021;21(4):1223. Available from: http://dx.doi.org/10.3390/s21041223
Ueda H, Abe R, Ohashi H, Iijima I, Ihara M, Takagi H, Hohsaka T. “Quenchbodies”: quench-based antibody probes that show antigen-dependent fluorescence. J Am Chem Soc [Internet]. 2011;133(43):17386–94. Available from: http://dx.doi.org/10.1021/ja205925j
Ueda H, Zhao S, Dong J, Jeong H-J, Okumura K. Rapid detection of the neonicotinoid insecticide imidacloprid using a quenchbody assay. Anal Bioanal Chem [Internet]. 2018;410(17):4219–26. Available from: http://dx.doi.org/10.1007/s00216-018-1074-y
Ueda H, Zhu B, Nosaka N, Kanamaru S, Dong J, Dai Y, Inoue A, Yang Y, Kobayashi K, Kitaguchi T, Iwasaki H, Koike R, Wakabayshi K. Rapid and sensitive SARS-CoV-2 detection using a homogeneous fluorescent immunosensor Quenchbody with crowding agents. Analyst [Internet]. 2022;147(22):4971–9. Available from: http://dx.doi.org/10.1039/d2an01051h
Zachary Kazaz—McMaster Integrated Science Program, Specialization in Biology 2025
Recent advancements in genome editing have facilitated major developments in both existing and new gene therapies (Hirakawa et al., 2020). The culmination of these findings became most evident on July 5th 2018, when Dr. Carl June and Professor Michel Sadelain published a seminal review article in the New England Journal of Medicine, reporting the clinical success of chimeric antigen receptor (CAR) T cell therapy against certain haematological cancers; the therapy subsequently received FDA approval for lymphoma and leukaemia treatment (June & Sadelain, 2018). T cells were modified with artificial receptors that associated the recognition of target antigens with the signalling and immunological responses of the T cell (Ellis et al., 2021). Furthermore, genome editing was successfully used to remove immune checkpoints limiting T cell functionality and improving receptor efficacy (Ellis et al., 2021). The successes of CAR T cell therapies have extended the applications of immune cell reprogramming to a population less investigated thus far — the B cell.
Adaptive immune responses are facilitated by white blood cells, or lymphocytes. There are two classes of adaptive immune response: humoral (antibody responses) and cell-mediated responses, primarily enabled by two types of lymphocytes: B-cells and T-cells, respectively (Alberts et al., 2002). Humoral adaptive immunity produces antigen-specific antibodies via B-cells that identify pathogens by binding to their expressed antigens, stimulating cell-mediated immunity by signalling T-cells, cytokines, macrophages and chemical mediators which attack and neutralize the pathogen (Johnson et al., 2018). Thus, B-cells regulate the entire adaptive immune system via general and specific mechanisms, facilitated by cytokine secretion and antigen presentation respectively (Rogers & Cannon, 2021). As a result, B-cells provide a means of modifying holistic immune responses which is a more effective immunotherapy than the sole modification of cell-mediated immunity found in current immunotherapies (e.g., T-cells).
Following the first, primary, response to a specific antigen, the responding, naive, B-cells will proliferate into a colony, differentiate into effector B-cells which produce antibodies, and following infection, will form memory B-cells which encode for and maintain the antigen-specific antibody of the pathogen encountered (Akkaya et al., 2019). This enables more rapid secondary active immune response mobilization if the same antigen exposure occurs subsequently, and can provide lifelong immune surveillance (Akkaya et al., 2019). As a result, humoral immunity facilitated by B-cells is the most capable mechanism of providing prolonged immune surveillance (Rogers & Cannon, 2021).
B-cells possess many unique characteristics that make them a promising focal point for genetic engineering in immunotherapy settings. B-cells can be easily isolated in abundance from peripheral blood, then subsequently activated, grown, and matured in culture ex-vivo (Liebig et al., 2009). It is possible to engineer these isolated B-cells to express a particular gene, mature them into effector B-cells, which produce large amounts of antibodies, and add them back to the host as a remarkably effective form of gene therapy enabling long-term immunity to disease, as seen in Figure 1 (Johnson et al., 2018). In particular, gene editing can be used to alter antigen specificity with a predetermined antibody by means that permit access to all functions of the B-cell during every stage of its life cycle (Rogers & Cannon, 2021).
Figure 1. Diagram displaying the procedure of B-cell immunotherapy in patients using host donor cells (Traxinger, 2019).
By altering the variations of modified antigen-specificity, the response of B-cells to target antigens, their subsequent production of antibodies, expansion, and formation of long-term immunity, we can introduce and evolve long-term antibody specificity to reprogram B-cells that will continue to adapt to their associated pathogens (Rogers & Cannon, 2021). This is particularly relevant, as a majority of current literature on B-cell engineering focuses on producing HIV-specific B-cells (Rogers & Cannon, 2021). The prolonged antibody expression of B-cell therapy can be used to suppress chronic viral infections such as HIV. More importantly, highly mutagenic viruses, such as HIV, are capable of evading antigen-specific antibody responses, and edited B-cells can adapt to these changes, enabling B-cells to keep pace with viral mutations (Ouyang et al., 2017). This is not possible with current therapies that use fixed antigen-specificities.
The broad array of B-cell functions open many potential immune cell gene therapies. For example, the ability of antibody production in effector B-cells allows them to be used as a long-term source for the production of therapeutic proteins in-vivo, such as bnAbs and factor IX to combat hepatitis C virus and HIV (Kuhlmann et al., 2018). Also, it is possible to create these “cellular factories” using stem cells or editing B-cells ex-vivo which can be altered with a variety of methods such as CRISPR/CAS9 and viral vectors which transport the modified gene to loci of interest (Luo et al., 2020). Moreover, naive B-cells can be modified to present antigens and suppress or prevent immune responses to particular antigens, which can be used to combat autoimmune diseases (Scott, 2011).
Interestingly, B-cell engineering offers potential as an improved means of vaccination. In May of 2019, Howell et al. published findings in Scientific Immunology showing that B-cells edited to express antibodies countering respiratory syncytial virus (RSV) produced highly effective and long-lasting protection from RSV infection in mice (Moffett et al., 2019). Such findings convey that sterilizing immunity to pathogens can be accomplished in a more effective manner than current vaccination methods are able to induce or sustain.
Though great strides have been made, moving forwards, many further steps are needed for B-cell therapies to become viable in the future. Up to now, studies have used mice, however, larger animal models more related to human immunology are required to gain more practical insights. Further, studies must be conducted long-term to assess the durability of antibody production and memory in edited B-cells (Rogers & Cannon, 2021). Additionally, novel methods of delivering B-cell therapies to patients must be developed to make B-cells commercially viable and accessible on a broad scale. Engineered B-cells should be manufactured en masse from universally matching batches of unrelated donor cells (i.e. allogeneic) instead of an exclusive therapy manfactured from a single sample of patient-related donor cells (i.e. autologous). Current studies propose using B-cells from induced pluripotent stem cells, which is currently being investigated for use in CAR T cells (French et al., 2015).
Today, genetically engineered B-cells present a great capacity as a highly effective immunotherapy for particular autoimmune disorders, cancers, chronic infectious diseases, and pathogens. Further experimentation is required to progress B-cell gene therapy to human trials, however, in the near future, we are likely to see B-cells accompany CAR T-cells as a novel immune cell therapy. In the far future, the broad and holistic applications of engineered B-cells may become the basis of future immunotherapies, vaccines, and disease treatments.
References
Akkaya M, Kwak K, Pierce SK. B cell memory: Building two walls of protection against pathogens. Nature Reviews Immunology [Internet]. 2019Dec13 [cited 2022Nov29];20(4):229–38. Available from: https://www.nature.com/articles/s41577-019-0244-2
Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Chapter 24: The Adaptive Immune System. In: Molecular Biology of the Cell [Internet]. 4th ed. New York, NY: Garland Science; 2002 [cited 2022Nov28]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK21070/
Ellis GI, Sheppard NC, Riley JL. Genetic engineering of T cells for immunotherapy. Nature Reviews Genetics. 2021Feb18;22(7):427–47. French A, Yang C-T, Taylor S, Watt SM, Carpenter L. Human induced pluripotent stem cell-derived B lymphocytes express SIGM and can be generated via a hemogenic endothelium intermediate. Stem Cells and Development [Internet]. 2015May [cited 2022Dec8];24(9):1082–95. Available from: https://www.liebertpub.com/doi/10.1089/scd.2014.0318
Johnson MJ, Laoharawee K, Lahr WS, Webber BR, Moriarity BS. Engineering of primary human B cells with CRISPR/Cas9 targeted nuclease. Scientific Reports [Internet]. 2018Aug14 [cited 2022Nov29];8(1). Available from: https://www.nature.com/articles/s41598-018-30358-0
June CH, Sadelain M. Chimeric antigen receptor therapy. New England Journal of Medicine. 2018Jul5;379(1):64–73. Kuhlmann A-S, Haworth KG, Barber-Axthelm IM, Ironside C, Giese MA, Peterson CW, et al.
Liebig TM, Fiedler A, Zoghi S, Shimabukuro-Vornhagen A, von Bergwelt-Baildon MS. Generation of human CD40-activated B cells. Journal of Visualized Experiments [Internet]. 2009Oct16 [cited 2022Dec4];(32). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3164064/
Luo B, Zhan Y, Luo M, Dong H, Liu J, Lin Y, et al. Engineering of α-PD-1 antibody-expressing long-lived plasma cells by CRISPR/Cas9-mediated targeted gene integration. Cell Death and Disease [Internet]. 2020Nov12 [cited 2022Dec6];11(973). Available from: https://www.nature.com/articles/s41419-020-03187-1
Moffett HF, Harms CK, Fitzpatrick KS, Tooley MR, Boonyaratanakornkit J, Taylor JJ. B cells engineered to express pathogen-specific antibodies protect against infection. Science Immunology [Internet]. 2019May17 [cited 2022Dec4];4(35). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6913193/
Ouyang Y, Yin Q, Li W, Li Z, Kong D, Wu Y, et al. Escape from humoral immunity is associated with treatment failure in HIV-1-infected patients receiving long-term antiretroviral therapy. Scientific Reports [Internet]. 2017Jul24 [cited 2022Dec4];7(1). Available from: https://www.nature.com/articles/s41598-017-05594-5
Despite the tremendous medical and technological advancements achieved in modern human history, only one disease has been officially eradicated from the world – smallpox (1). Nevertheless, another life-threatening disease came painstakingly close to being eliminated. Polio, or poliomyelitis, is a disease caused by the wildtype poliovirus (colloquially referred to as WPV) (2). In this case, wild type refers to the naturally occurring virus – a distinction that becomes important later. WPV is commonly transmitted through contact with contaminated feces and infection commonly occurs in children under the age of 5 (3,4). Approximately 75% of people infected with the poliovirus show no symptoms, while a little less than 25% experience mild symptoms like fatigue, fever, muscle pain, and stiffness (1,3,5). In the most severe cases, 1 in 200 become paralyzed while 2-10% of those paralyzed by polio die because it immobilizes the muscles needed for breathing (5,6).
SOURCE: Shutterstock
Across 125 endemic countries, the disease had paralyzed 350,000 children annually up until 1988 when the World Health Assembly launched the Global Polio Eradication Initiative (GPEI) (5). This vaccination program was on a mission to globally eradicate all cases of polio by raising funds to implement national immunization programs in nations that could not stop the transmission of the virus on their own (7). Two different types of vaccines were used. First, an injectable vaccine that contained the inactive form of the poliovirus (abbreviated to IPV), and an oral vaccine with a live, but weakened, form of the virus (OPV). The OPV was pivotal for preventing transmission in endemic areas with unsanitary conditions because it causes the virus to replicate in the gut and build immunity in the intestines. This then prevents it from being shed through feces into the environment (1). It’s also cheap to make and easy to administer on a community-wide level since there are no injections necessary (2). On the other hand, IPV, which builds immunity in the bloodstream and prevents the virus from causing paralysis, is useful in extremely sanitary areas like North America because children there have lower immunity to the virus (1).
SOURCE: Sebastian Meyer/WHO Iraq
For the most part, the GPEI had succeeded. WPV cases declined by over 99% since 1988, with only 175 cases reported in 2019 (5). The WHO had certified the global eradication of 2 out of the 3 wildtype strains in 2015 and 2019 (8).
So, how come polio hasn’t joined smallpox on the list of fully eradicated diseases? Well, there has been a recent and concerning phenomenon where the virus in the OPV is being transmitted from a vaccinated host to a susceptible person with no immunity to polio (6). This is a rare occurrence, about 1 in 2.5 million cases, yet it was common enough to spark outbreaks of vaccine-derived poliovirus (VDPV) in about 33 countries as of 2020 (2). More recently, wastewater studies in London and throughout the state of New York showed circulating cases of VDPV in July 2022 (5).
The problem here is two-pronged: there is not enough immunization coverage in these areas, and the vaccines being administered don’t prevent outbreaks. The WHO recommends a vaccination coverage of more than 95% against polio, yet they estimate that the coverage in the UK and US were 93% and 92% respectively (5). Some of the VDPV outbreaks occurred in areas with inadequate coverage. Unvaccinated people are at risk of having the weakened virus replicate in their gut and undergo mutations that revert it back to a harmful form (2). Second, IPVs are primarily used in these regions over OPVs after the latter was withdrawn from routine immunizations in 2016 (8). IPVs can prevent the virus from becoming paralytic in the host, but they do not prevent its transmission to those who are more susceptible (1).
Currently, researchers are at odds with each other on how to solve this health crisis. Some suggest reintroducing OPVs to all regions to increase coverage and prevent transmission (1), while others want to discontinue its use and shift towards the more resource-intensive IPV to mitigate additional VDPV infections (2,4). One silver lining is that by arriving on the cusp of another contentious vaccination topic, we’re primed to make advancements in these difficult, yet crucial conversations.
4. Duintjer Tebbens RJ, Thompson KM. Polio endgame risks and the possibility of restarting the use of oral poliovirus vaccine. Expert Rev Vaccines. 2018 Aug 9;17(8):739–51.
5. WHO. Detection of circulating vaccine derived polio virus 2 (cVDPV2) in environmental samples– the United Kingdom of Great Britain and Northern Ireland and the United States of America [Internet]. World Health Organization. 2022 [cited 2022 Nov 25]. Available from: https://www.who.int/emergencies/disease-outbreak-news/item/2022-DON408
Chances are you’ve heard mentions of a COVID-19 pill, but you may be wondering, “What actually is it?” There are many emerging treatments for COVID-19 meant for people who are already infected with the virus, including treatments taken orally. These treatments differ from vaccinations because they actively treat the virus whereas vaccines are used as a preventative measure to avoid getting infected in the first place. Similarly to the COVID-19 vaccines, however, Pfizer is one of the first to hop onto the COVID-19 pill train, having created the first FDA-approved oral treatment for COVID-19, Paxlovid (1).
What exactly is Paxlovid?
Paxlovid is the combination of two nirmatrelvir pills and one ritonavir pill, all taken twice a day over the span of five days (2). Nirmatrelvir, a drug created by Pfizer, is the drug in Paxlovid that contains Paxlovid’s antiviral properties, limiting the replication of the virus (2). Ritonavir, an existing drug typically used in the treatment of HIV/AIDS among other things, allows nirmatrelvir to remain in the body at higher concentrations for longer periods of time; it does this by being a CYP, cytochrome P450, inhibitor (3). CYPs are enzymes that are involved in the termination of many drugs, including nirmatrelvir, so inhibiting them enhances and prolongs the antiviral properties of nirmatrelvir. It is recommended that Paxlovid is taken within five days of symptom onset (1).
SOURCE: CBC News
Outcomes
Nirmatrelvir and ritonavir clearly work together to create a powerhouse of a COVID-19 treatment that demonstrates immense benefits. A nearly five-month-long clinical trial conducted by Pfizer, which concluded in December 2021, showed an 89% decrease in severe illness and death when the drug was taken within three days of symptom onset as compared to a placebo (4). This trial, called EPIC-HR was done on adults who present a high risk of COVID-19 progressing to a severe illness. In the same trial, 0.7% of patients who received Paxlovid were hospitalized as opposed to 6.5% who received the placebo being hospitalized or dying (4). In a second trial, EPIC-SR, done on adults at a more standard risk, hospitalization was reduced by 70% as compared to the placebo (4).
However, Paxlovid is not perfect. It’s not recommended for certain populations such as those with severe kidney or liver impairments (1). A lack of research also makes it difficult to prescribe to certain populations such as people under 40 kilograms, pregnant or lactating people, and those on drugs that could have potentially dangerous interactions with Paxlovid (5).
Implications and the Future
Paxlovid has not only paved the way for other COVID-19 antiviral pills, but also for other forms of COVID-19 treatments. These include antiviral-type treatments, such as the oral treatments Paxlovid and Molnupiravir, and the intravenous treatment Remdesivir (6). Monoclonal antibodies are another form of COVID-19 treatment that improve the immune system’s response to the virus as opposed to targeting the actual virus itself. Bebtelovimab is a monoclonal antibody that combats COVID-19 through intravenous injection (6). Paxlovid is just the beginning, and the creation of new COVID-19 treatments can improve the accessibility of a treatment for those who can not take Paxlovid for a multitude of reasons or in situations where Paxlovid is not available.
SOURCE: CDC
The accessibility of Paxlovid has actually been a large issue. This is in part due to the lack of transparency that producers of Paxlovid have demonstrated, particularly in the realm of costs and remaining supply. The lack of transparency has led to challenges in lower-income countries receiving the treatments, resulting in the WHO declaring its concerns regarding Paxlovid accessibility (5). The COVID-19 pandemic has already led to great global divides, and Paxlovid’s limited accessibility gives it the potential to further increase disparities within global health.
Although the future of COVID-19 treatments and the potentially negative implications of Paxlovid are greatly unknown, its introduction into the world of healthcare is quite beneficial and exciting. Paxlovid has opened up a new realm of research within the topic of COVID-19 and has provided an amazing opportunity for collaboration to lead to many incredible discoveries!
References
Louisiana Department of Health. FDA authorizes first antiviral pills for COVID-19 [Internet]. [cited 2022 Nov 28]. Available from:https://ldh.la.gov/news/paxlovid
Cancer is a leading cause of death globally, accounting for nearly 10 million deaths in 2020, or nearly one in six deaths1. Cancer results from the uncontrollable division of abnormal cells. The most common cancer are breast, lung, colon and rectum, and prostate cancer1. While the survival rate varies depending upon the types of cancer, the chance of survival is significantly greater when cancer is diagnosed earlier. In general, the rate of survival of cancer is 91% when diagnosed at an early stage, and 26% when diagnosed at a later stage7. However, recently scientists have proposed the use of blood tests to diagnose various types of cancer. Blood testing is quick and non-invasive, relatively inexpensive, and readily available. Currently available cancer blood tests include Complete blood count (CBC), CancerSEEK Test, Galleri multicancer early detection (MCED) test and PanSeer Test.
Complete blood count (CBC)
SOURCE: Verywell Health
CBC is a common blood test used to detect a variety of disorders. The results of the CBC can be used to direct one’s diagnosis towards a particular disease2. Moreover, this test can be used to detect Blood Cancers, such as leukemia and lymphoma3.
CancerSEEK Test
SOURCE: Dr. Lal PathLabs Blog
The CancerSEEK Test detects cell-free DNA, cfDNA, and identifies eight biomarkers released by Tumour cells4. This blood test can detect the problems in the early stage of tumor5. Tumors are detected by mutations in genomic positions, such as substitutions, insertions and deletions4. So far, eight types of cancer can be detected with over 99% accuracy, including ovarian, liver, stomach, pancreatic, esophageal, colorectal, breast, and lung4.
Galleri multicancer early detection (MCED) test
SOURCE: Johns Hopkins Medicine Newsroom
The Galleri MCED is a novel, high-performance genomic technology that can detect signals from Cancerous cells at the early stage6. The Galleri MCED test aims to identify cfDNA circulating in the blood, and specifically recognized DNA methylation. The test result can indicate the specific type of cancer, and identify in which organ cancerous cells are present4. There are 12 types of cancer at the early stage that can be detected with 93% accuracy, including anorectal, colorectal, esophageal, gastric, head and neck, hormone receptor-positive breast, liver, lung, ovarian, and pancreatic cancers, in addition to multiple myeloma and lymphoid neoplasms4.
PanSeer Test
SOURCE: Galleri for HCPS
The PanSeer test uses a similar approach to the Galleri MCED test. This test was developed by the Taizhou Longitudinal Study that compared blood samples of individuals with and without cancer. In this study, they compared approximately 400 blood samples with cancer and approximately 400 blood samples without cancer. After that, they record physical measurements and questionnaires about the cancer occurrence, collecting affectional plasma and tissue samples at 3-year intervals7. The test detected patterns of DNA methylation, and 95% of asymptomatic individuals were diagnosed with cancer by using standard detection methods4. However, this test is unable to determine the exact location of the cancer4.
Overall, although blood tests can be used to detect cancer at the early stage which increases the rate of survival, not all types of cancer can be detected. In addition, some confounding factors, such as personal lifestyle, environmental pollution, and individual genetic composition, would make it difficult to develop standardized blood tests for all individuals7.
7. Chen X, Gole J, Gore A, He Q, Lu M, Min J, et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test [Internet]. Nature News. Nature Publishing Group; 2020 [cited 2023Jan2]. Available from: https://www.nature.com/articles/s41467-020-17316-z
Daniel Constantinescu—McMaster Health Sciences 2026
The field of bioacoustics is uncovering amazing new ways to study the natural world. It involves investigating the production, transmission, and reception of animal sounds (1). Bioacoustics can be used to assess habitat health, species diversity, wildlife behaviour, and more (2). Audio has the incredible capability of recording species in very large areas, while video is limited by a camera’s field of view (3). Given that commercial audio equipment costs from $500 to $1000 per recorder, using audio for studies has not usually been a viable option in the past. Today; however, the price of recording equipment has decreased drastically.
SOURCE: Noble Research Institute
After the collection of the audio, artificial intelligence algorithms can be used to analyze the recordings. Thousands of hours of audio can be examined rapidly to identify different species, and collect information about them. One can only imagine the benefits of bioacoustics, especially when acquiring data for species such as birds, monkeys, or bats, which are either constantly on the move, or nocturnal. Additionally, this method of research means that humans can spend less time in potentially dangerous environments (1). To continue, audio can contain emotional information because the vocalizations of some species differ during positive and negative experiences (1). As such, scientists can use bioacoustics to find out if certain human activities, such as shipping, or seismic surveys affect certain animals. These assessments can be particularly useful in large-scale farming. Most research to date has been focusing on reducing the negative experiences for animals, but the concept of animal welfare has evolved to ensure that they have positive experiences as well. Emotions can sometimes be deciphered by analyzing the frequency of the sounds that an animal makes. Next, another benefit of bioacoustics is the ability to automatically infer individuality information about animals, with a particular study achieving 71% accuracy in this task (1). Being able to recognize specific animals will make estimating the number of species in a population much more reliable.
SOUND: Yale Environment
There are also interesting techniques being developed in marine science that use bioacoustics. Passive acoustic sonar, for example, is a method of detecting the location of an animal using sound. Multiple microphones are set up, evenly spaced, and the difference in the time taken for the vocalization to reach each microphone can be used for triangulation (1). A challenge that is faced in the field; however, is the need to label sounds that occur in a recording manually. This can take a very long time. That being said, this problem is being addressed with the development of programs that can find the regions of interest in audio on their own. To conclude, it is clear that the field of bioacoustics demonstrates many promising features. In the near future, audio could become a common data collection method, helping scientists make meaningful decisions to help the environment.
References
1. Mcloughlin MP, Stewart R, McElligott AG. Automated bioacoustics: methods in ecology and conservation and their potential for animal welfare monitoring. J R Soc Interface. 2019 Jun 28;16(155):20190225.
Cancer refers to any one of a large number of diseases characterized by the development of abnormal cells that divide uncontrollably and have the ability to infiltrate and destroy normal body tissue as well as spread throughout your body.1 It is the second leading cause of death in the world.1 Cancer is caused by mutations to the DNA within cells. These mutations can instruct a healthy cell to allow rapid growth, fail to stop uncontrolled growth or make mistakes when repairing DNA errors.1
Cancer is a highly adaptable disease which causes it to endure the constantly changing microenvironments that its cells encounter2. Cancer cells have a certain degree of adaptive immune resistance, a process in which the cells change their phenotype in response to cytotoxic or proinflammatory immune response9. This response is triggered by the recognition of cancer cells by T cells, leading to the production of immune-activating cytokines9. Cancer cells then hijack mechanisms developed to limit inflammatory and immune responses and protect themselves from the T cell attack9. Because of this process, cancer continues to thwart patients, researchers, and clinicians despite significant progress in understanding its biological underpinnings.3
SOURCE: Future Processing Better Future
As more is learned about the disease itself, more can be learned about how tools can be useful in treatment plans. Currently, artificial intelligence is used in the detection of cancer as it effectively analyzes complex data from many modalities, including clinical text, genomic, metabolomic, and radiomic data (the extraction of mineable data from medical imaging)6. An example of artificial intelligence in cancer diagnosis is imaging tests, which allow your doctor to examine your bones and internal organs in a non-invasive way7. This may include a computerized tomography (CT) scan, bone scan, magnetic resonance imaging (MRI), positron emission tomography (PET) scan, ultrasound and X-ray7.
SOURCE: Stanford Medicine
Artificial intelligence is also used for cancer treatment through something called “precision medicine.” Precision medicine uses specific information about a person’s tumor to help make a diagnosis, plan treatment, and/ or evaluate the effectiveness of treatment5. It involves testing DNA from a patient’s tumor to identify the mutations or other genetic changes that drive their cancer8. Doctors can select a treatment plan that is best suited for that specific patient. Because no two cancers are identical, precision medicine is important as each patient has a unique combination of genetic changes2.
Overall, artificial intelligence provides a gateway to push the boundary of cancer treatment. Currently, it is used most in the detection of cancers through CT scans, bone scans, and PET scans, among others. However, as artificial intelligence is adopted into clinical oncology, its potential to redefine cancer treatment is becoming evident.
3. Linda WB, Hosney A, Schabath MB, Giger ML, Birkbak N, et al. Artificial Intelligence in cancer imaging … – wiley online library [Internet]. American Cancer Society. John Wiley & Sons; 2019 [cited 2022Nov13]. Available from: https://acsjournals.onlinelibrary.wiley.com/doi/full/10.3322/caac.21552
6. Hunter B, Hindocha S, Lee RW. The role of artificial intelligence in early cancer diagnosis [Internet]. National Library of Medicine. U.S. National Library of Medicine; 2022 [cited 2022Nov15]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8946688/
9. Ribas A. Adaptive immune resistance: How cancer protects from immune attack [Internet]. National Library of Medicine. U.S. National Library of Medicine; 2015 [cited 2022Nov25]. Available from: https://pubmed.ncbi.nlm.nih.gov/26272491/