Journal 1: As a senior in high school, I have gradually gotten to understand that it is really important to be involved in activities outside the classroom that are respective to my future academic goals in college. My interest in biomedical engineering thus leads me to try and find related experiences that will not only give knowledge but also give richness in understanding within the field. I spend my free time editing research papers, literature reviews, and other scholarly articles as part of the effort to build my analytic skills and stay in tune with ongoing discussions in scientific circles. I am also very interested in learning to play new musical instruments and languages, striving to achieve fluency both musically and linguistically. In summers, the idea is to get hands-on experience by interning in academic labs. Last summer, I had the opportunity to intern with the Dinolfo Group at RPI and work on Stern–Volmer quenching, measuring the quenching constants of various quenchers on specific photocatalysts used in photoredox reactions. This experience greatly enhanced my comprehension of chemical processes and experimental techniques. Another major commitment of mine outside of school is Science Olympiad. It is a club in which schools compete with their students in various events that require both knowledge and engineering. Since then, this club has polished my teamwork abilities and problem-solving skills. It's an area where I can apply my love for science in a competitive, yet collaborative manner. I don't stop at academics; there are more things that interest me. I'm an avid lover of the board game 五子棋; it is a strategic mix between checkers and Connect Four, said to have been created around Japan's Edo period. I love snowboarding, ice skating, and soccer. Interestingly, I am multilingual; my first three languages are English, Patois, and Spanish, then French, Malaysian Malay, Indonesian Malay, Cantonese, Korean, Vietnamese, and Arabic.
Journal 2: My relationship with grades has evolved over time. Initially, grades were a way to measure my understanding of the material, but as I’ve matured, I’ve come to see them as just one aspect of my academic journey. While I do strive for high grades, they are not my sole focus. Instead, I concentrate on gaining a deeper understanding of the subjects I am passionate about, particularly in biomedical engineering. My motivation comes from a genuine curiosity and a desire to contribute meaningfully to the scientific community. Grades are important, but they are a byproduct of my dedication to learning rather than the ultimate goal. One of my favorite memories inside the classroom was when we were learning about complex physical phenomena (light being seen as a wave and a photon) in my AP Physics class. The teacher made the abstract concepts come alive with engaging demonstrations that illustrated the principles we were studying. It was in these moments that I realized my love for science and how much I enjoy the process of discovery. Outside the classroom, my favorite memory is from Science Olympiad. Spending months studying and leading the team into the state tournament made it an unforgettable experience. It was not just about the competition but also about applying our knowledge in creative and practical ways. What I love most about school is the opportunity to learn and explore new ideas. Whether it’s delving into a complex scientific theory or learning a new language, intellectual stimulation is something I truly cherish. However, what I find less appealing is the pressure that sometimes comes with the pursuit of high grades. It can detract from the joy of learning, turning education into a race rather than an enriching experience. E=mc² attracted me because of its reputation as a place where intellectually curious students can thrive. I’ve heard that it fosters an environment of collaboration and creativity, which aligns perfectly with my academic and personal interests. The opportunity to be surrounded by like-minded individuals who share my passion for learning is incredibly appealing. I am excited to contribute to and grow within such a vibrant community.
Journal 3: I'm deeply interested in the intersection of pharmacogenomics and biomedical engineering because it embodies the fusion of personalized medicine and cutting-edge technology—two areas that have always captivated me. This field has the potential to revolutionize how we approach healthcare, making treatments more effective and tailored to individual genetic profiles. My fascination with how tiny variations in DNA can influence drug responses drives my desire to delve deeper into this topic, particularly as it aligns with my broader goal of advancing biomedical technology. From my previous research, I understand that pharmacogenomics involves the study of how genes affect a person’s response to drugs. It’s a field that combines pharmacology and genomics to develop effective, safe medications and doses tailored to a person’s genetic makeup. Biomedical engineering, on the other hand, applies principles of engineering to solve biological and medical problems, including the development of devices, systems, and software. The integration of these two fields could lead to innovations such as personalized drug delivery systems or genetically informed medical devices. Last year, my research focused on the efficacy of biomaterials used in biomedically engineered pancreases. This year, I aim to build on that knowledge by exploring how pharmacogenomic data can inform the design of biomedical devices and technologies. This extension will allow me to understand not just the biochemical interactions at play but also how engineering principles can be applied to create more personalized medical solutions. Five questions I have as I begin my research are: How can pharmacogenomic data be integrated into existing biomedical technologies? What challenges exist in designing devices that can adapt to a patient’s genetic profile? How does the variability in genetic data impact the development of universally applicable biomedical devices? What are the ethical considerations in using genetic information to design personalized medical technologies? How can biomedical engineering innovations improve the accessibility and affordability of personalized medicine? Three people I can think of that could support me are my PI at RPI, my aunt who works in law surrounding medical ethics, and the biotechnology teacher here at Guilderland.
Journal 4: I think I know that pharmacogenomics is about how people’s genetic makeup influences their response to drugs. I’ve heard that some people metabolize drugs faster than others, which can affect how effective a medication is or whether it causes side effects. I think I know that biomedical engineering might play a role in this by creating devices or systems that help deliver the right dose of a drug based on a person’s genetic profile. I think the combination of pharmacogenomics and biomedical engineering could help make personalized medicine a reality. I also think I know that pharmacogenomics is a growing field because personalized treatment is becoming more important in healthcare. The idea is that instead of prescribing the same medicine to everyone, doctors could look at your DNA and determine the best treatment for you. I’m not entirely sure, but I think biomedical engineers might develop technologies to analyze genetic data more efficiently or design devices that interact with patients in ways that are informed by their genetic information. I think I know there are ethical concerns about using genetic data in medicine, like privacy and the potential for discrimination.
I know I know that pharmacogenomics focuses on how variations in genes, like those in the CYP450 enzyme family, can significantly alter how individuals metabolize drugs, leading to variations in drug efficacy and risk of side effects (U.S. National Library of Medicine). For example, genetic differences in the CYP2D6 gene can cause people to metabolize opioids or antidepressants too quickly or too slowly, impacting their therapeutic effects (National Institutes of Health). Biomedical engineering, on the other hand, applies engineering principles to solve medical problems, often through the development of medical devices, imaging tools, and drug delivery systems (National Institute of Biomedical Imaging and Bioengineering). This field can create personalized medical devices or systems that interact with genetic information to ensure that drugs are delivered at the right dose and pace (Parvez & Kang, 2020). I also know that personalized medicine uses pharmacogenomic data to tailor treatments for individuals, increasing the likelihood of success while reducing adverse effects (Relling & Evans, 2015). The integration of pharmacogenomics and biomedical engineering could push healthcare toward a future where treatments are highly individualized (Manolio et al., 2019).
Journal 5:
I don't know's:
- I don’t know if pharmacogenomics is currently widely implemented in drug development.
- I don’t know the extent to which biomedical engineering has contributed to advancements in personalized medicine.
- I don’t know how different genetic variations specifically affect drug efficacy in diverse populations.
- I don’t know if there are specific ethical concerns related to tailoring medicines based on genetic profiles.
- I don’t know the long-term impacts of using pharmacogenomics in clinical practice.
- I don’t know how biomedical engineering can improve the delivery mechanisms for personalized drugs.
- I don’t know the regulatory challenges in approving pharmacogenomic-based medicines.
- I don’t know if the cost of developing pharmacogenomic-based drugs is significantly higher than traditional methods.
- I don’t know what advancements have been made in using biomedical devices for drug administration in targeted ways.
- I don’t know if pharmacogenomics could reduce adverse
Need to know:
Understanding how genetic variations impact drug efficacy is crucial for the development of better medicines. The way individuals metabolize and respond to drugs can vary significantly based on their genetic makeup, and this knowledge can help inform the creation of more effective, targeted therapies. This is particularly important for diverse populations, as a one-size-fits-all approach to medicine can lead to inefficiencies and adverse reactions in certain groups. Investigating this topic will not only contribute to the advancement of personalized medicine but also help address disparities in healthcare, ensuring that treatments are more inclusive and effective across different demographic groups.
Journal 6:
Need to Know Question:
How do genetic variations affect drug efficacy in diverse populations?
To address this question, understanding the role of genetic differences in drug metabolism and response is critical, especially given the push toward personalized medicine. Genetic variations, known as polymorphisms, can significantly alter the way individuals process drugs, leading to variations in efficacy and safety. Two reliable sources—one a scientific study and the other a review article—will provide evidence to explore how these variations impact drug efficacy across diverse populations.
In a review of pharmacogenomics and its role in drug therapy, an article from Nature Reviews Drug Discovery highlights the importance of genetic differences in enzymes responsible for drug metabolism, particularly those in the cytochrome P450 (CYP) family. CYP enzymes are key players in metabolizing many drugs, and genetic polymorphisms in these enzymes can lead to “poor,” “intermediate,” or “ultrarapid” metabolism in different individuals. For example, variations in the CYP2D6 gene affect the metabolism of antidepressants, antipsychotics, and opioids. In certain populations, such as those of African or Asian descent, specific alleles of CYP2D6 are more prevalent, meaning individuals in these groups may metabolize drugs more slowly or quickly compared to individuals of European descent. This can lead to differences in drug efficacy and risk of side effects. For instance, an ultrarapid metabolizer might break down a drug too quickly for it to have therapeutic effects, while a poor metabolizer could experience toxicity from drug accumulation .
The second source, a study published in The New England Journal of Medicine, examines how genetic differences in drug transporters can influence drug response. Transport proteins like P-glycoprotein (P-gp) regulate the absorption and distribution of many drugs, affecting their bioavailability. Polymorphisms in the genes encoding these transporters have been linked to variations in how drugs are absorbed and distributed in different populations. The study found that certain P-gp polymorphisms are more common in individuals of East Asian descent, influencing their response to drugs like statins, which are used to lower cholesterol levels. The study further emphasizes the importance of considering these genetic differences in clinical practice, as individuals with certain genetic profiles may require adjusted doses or alternative medications to achieve optimal therapeutic outcomes .
The evidence from these sources show the significant role that genetic variations play in drug metabolism and response, particularly in diverse populations. Genetic polymorphisms in drug-metabolizing enzymes and transport proteins can lead to significant differences in how individuals from different genetic backgrounds process medications. This highlights the importance of incorporating pharmacogenomics into drug development and clinical practice to ensure that treatments are tailored to the genetic profiles of diverse patient populations. By doing so, drug efficacy can be improved, adverse side effects can be minimized, and the leap to fully personalized medicine can be made.
Journal 7: October SDA Reflection:
The assignment's level of freedom was not completely limiting, but not completely free either, which I appreciated. The structure allowed me to engage with the subject without giving me too little instruction, allowing me to not become completely confused or stuck with what to do. The flexibility was definitely refreshing, as it gave me space to explore additional ideas without feeling constrained by rigid guidelines. However, the open-ended nature also came with its own challenges. Without a strict format, it can be difficult to know whether or not I was on the right track.
One positive aspect of the assignment was the opportunity to dive into the basics of pharmacogenomics and genetics in general. This let me kind of relearn the general parts of the subject but also explain my subject to those who may not be familiar. On the flip side, a negative would be that with such a vast amount of information on the subject, it was challenging to distill the key points correctly without leaving out important details.
I dedicated a solid few hours to the project. The research, planning, and actual crafting of the visual content required careful thought, especially ensuring that the scientific concepts were accurately communicated while being accessible to a broader audience. One key discovery I made was how essential pharmacogenomics is to the future of medicine. The variations in drug responses based on genetics showed me how much our genetic makeup influences our health beyond what I previously understood. On a personal level, I realized I enjoy breaking down complex scientific ideas into simpler, more digestible formats.
The most important thing I learned was the significance of understanding genetic diversity in healthcare. Recognizing how specific populations may respond differently to medications highlights the need for a more personalized approach to treatment, which could significantly reduce adverse drug reactions. This understanding showed the importance of inclusivity in medical research—ensuring all populations are represented to make treatments more effective for everyone.
When it comes to identifying a problem, I learned that sometimes, the issue is layered. It isn’t just about the science itself but also about how the science is applied—such as ensuring equitable access to personalized treatments. Looking back, I am proud of the way I was able to synthesize technical information into a more user-friendly format. However, I still need to work on how I manage time when facing projects with broad freedom.
Throughout the process, I consulted with Mrs. McTiernan, who guided me in refining my approach to the assignment. Her insights were invaluable in helping me strike a balance between scientific accuracy and accessibility. Moving forward, the information I’ve gained about pharmacogenomics will be useful in understanding how future innovations in medicine might benefit different populations.
As for my new essential question: How can personalized medicine and pharmacogenomics be integrated into mainstream healthcare to improve treatment outcomes for diverse populations?
Journal 8:
My newer "need to know" question would be: How does one incorporate personalized medicine and pharmacogenomics into mainstream healthcare in order to elicit better treatment outcomes across diverse populations? I think this question falls under the topic of the HOTQs because I am looking to suggest another or better healthcare model based on personalized medicine and pharmacogenomics. The wording itself, such as "How would you improve...?", and "What way would you design...?" since I try to build on a view on how I can integrate those concepts into the healthcare systems of the world. To answer this question I'll have to put together knowledge from my pharmacy class along with health policy and patient care.
Some sub-questions are: What are the main barriers to the widespread adoption of pharmacogenomics in healthcare systems? This sub-question requires me to consider and categorize the various obstacles-financial, technological, and educational-which in fact create a barrier to this use. What policy changes could be instituted to support wider implementation of personalized medicine? This sub-question leads me to assess how the policies that have so far been set up are working and if any new regulations should be instituted or revised. It requires critical judgement for what policy changes in healthcare would yield the best results in patient care through personalized medicine. And lastly, how could healthcare professionals be educated and trained to effectively implement pharmacogenomics in clinical practice? This falls under the application domain of HOTQs. Focusing on how to apply educational strategies to improve healthcare as a whole and asking what methods can be used to improve knowledge.
I believe the best sub-question to help is, "What are the main barriers to the widespread adoption of pharmacogenomics in healthcare systems?" This question allows me to analyze the main challenge in pharmacogenomics: ethics. By understanding the barriers, it would be easier to find solutions and more practical ways to implement personalized medicine that can make everyone involved happy.
The first resource I plan on using is someone I've read a lot of: Magnus Ingelman-Sundberg.
Ingelman-Sundberg, Magnus, et al. "Pharmacogenomic Biomarkers for Improved Drug Therapy—Recent Progress and Future Developments." AAPS Journal, vol. 22, no. 3, 2020.
This article provides a huge overview of all the (relatively) current going ons for pharmacogenomics.
The second resource I plan on using is a bioethics based one:
Johnson, Julie A., and Relling, Mary V. "Pharmacogenetics: Ethical Issues and Challenges in Implementing Genomic Medicine." Nature Reviews Genetics, vol. 13, no. 1, 2012.
This source will allow me to analyze the various arguments behind gene editing in general and personalized medicine.
The third source is one that would allow me to review the potential for pharmacogenomics to reduce adverse drug reactions, a great example of the impact of this field as a whole.
Philips, K. A., et al. "Potential Role of Pharmacogenomics in Reducing Adverse Drug Reactions: A Systematic Review." JAMA, vol. 286, no. 18, 2001, pp. 2270-2279
Journal 9:
After looking further into the sources I gathered on pharmacogenomics and personalized medicine, I came away with insights that not only addressed my main question, but also highlighted the complexities and challenges standing in the way. It’s clear that pharmacogenomics has incredible potential to transform medicine by tailoring drug treatments to a patient’s genetic profile. However, as I looked deeper, I realized just how difficult it is to implement this personalized approach on a broad scale.
One of the most compelling findings came from Ingelman-Sundberg’s work, which laid a solid foundation for understanding the practical impact of pharmacogenomics. They explain that “genetic polymorphisms in drug-metabolizing enzymes, particularly in the CYP450 family, significantly influence drug efficacy and the risk of adverse drug reactions” (Ingelman-Sundberg et al.). This means that certain genetic markers can guide doctors in adjusting medication dosages to avoid side effects or ineffective treatment. This research points to a future where people could potentially avoid harmful drug reactions simply because their body metabolizes a drug differently from the “average” patient.
However, as Ginsburg and Willard emphasize in Genomic and Personalized Medicine, there is currently “a lack of standardized protocols for the implementation of pharmacogenomics in clinical settings” (Ginsburg and Willard, 145). Different healthcare providers and institutions each have their own approaches to genetic testing, leading to inconsistent care and varied treatment outcomes. It made me realize that having the technology available is only part of the puzzle. Without a unified framework or guidelines, it’s hard to make pharmacogenomics a routine part of healthcare. This brings up the need for “established clinical guidelines that can bridge the gap between research and practical application” (Ginsburg and Willard, 147).
Another major issue that Johnson and Relling discuss is the ethical side of pharmacogenomics—particularly, the privacy concerns that arise when dealing with genetic information. They warn that “fears about misuse of genetic data could become a barrier to the adoption of pharmacogenomics, as patients may hesitate to undergo testing” (Johnson and Relling, 320). This reluctance isn’t trivial. It’s a roadblock that could impede pharmacogenomics’ widespread use. Addressing this challenge means creating strong privacy protections that inspire trust among patients. Johnson and Relling argue that “effective policies to safeguard genetic data must be part of any effort to integrate pharmacogenomics into clinical practice” (Johnson and Relling, 322).
Phillips et al. add to the urgency by providing a concrete example of how pharmacogenomics could benefit patient safety and reduce healthcare costs. They report that “adverse drug reactions (ADRs) account for nearly 6.7% of all hospital admissions” and suggest that many of these could be prevented through pharmacogenomic interventions (Phillips et al., 2131). This statistic made the benefits of personalized medicine feel even more tangible. Personalized medicine isn’t just about precision; it’s about safety. If we can prevent even a portion of these hospitalizations by using genetic data to guide treatment, the healthcare system could become safer and more cost-effective for everyone.
Despite these insights, there are still unanswered questions. One lingering issue is how healthcare systems could realistically adopt standardized pharmacogenomics protocols. What would those guidelines entail, and how can they be adapted to different healthcare settings? I also wonder what policies would encourage healthcare providers to invest in the infrastructure and training necessary for pharmacogenomics. Finally, I’m still curious about privacy concerns and how they can be addressed in a way that feels truly reassuring to patients.
To further explore these questions, I found two additional sources. The first is a paper by Manolio et al., titled “Implementing Genomic Medicine in the Clinic: The Future Is Here,” which discusses practical steps for bringing genomics into clinical settings while addressing privacy concerns. The second is an article by Collins and Varmus, “A New Initiative on Precision Medicine,” which looks into policy options that could support the adoption of pharmacogenomics on a larger scale. Both sources should help me dig deeper into the structural and ethical issues that still stand in the way of making pharmacogenomics a mainstream part of healthcare. I’m looking forward to seeing what these resources reveal and moving closer to answering my essential question. And, to clarify, I get access to most of these articles through institutional access from RPI and books around our lab.
Journal 10:
My essential questions matter because they address the potential of pharmacogenomics and personalized medicine to revolutionize healthcare. As society can move closer to more individualized treatments, understanding these concepts can lead to better health outcomes, fewer adverse drug reactions, and more efficient use of resources in the medical field. This is significant not only to patients but also to the healthcare system as a whole, as it may reduce costs and improve the quality of care.
These questions impact multiple groups: patients, healthcare providers, pharmaceutical companies, and researchers. Patients could experience fewer side effects and more effective treatments, healthcare providers would be able to offer more precise care, and pharmaceutical companies might see shifts in drug development priorities. Researchers studying genetics and pharmacology are also directly affected, as these questions can give rise to new areas of investigation.
The evidence supporting the importance of personalized medicine and pharmacogenomics come mostly from academic sources. For example, Johnson and Relling highlight the potential of pharmacogenectics to "tailor drug therapy to individual patients," thereby reducing adverse reactions. Philips et al. in JAMA emphasize that pharmacogenomics could be key in reducing the high rate of adverse drug reactions. These sources add some credibility to the argument that pharmacogenomics can meaningfully impact patient care and health outcomes.
My essential questions connect to several other fields, including genomics, biochemistry, biophysics, and policy. The ethical and societal implications of personalized medicine are also grounded in bioethics and legal studies, as questions of accessibility and fairness arise. Furthermore, this topic overlaps with health economics, given that personalized treatments may be expensive at first.
Initially, I assumed that personalized medicine was already widely implemented and accessible through compounding pharmacies, but my research has shown that it is still largely in the development phase and accessible only to certain populations. Additionally, I had assumed that genetic testing for drug response would usually give clear and easily interpretable results; however, I found that our genetic basis for drug responses is still incomplete, and many factors outside of genetics, like age, can influence how patients respond to medications.
For my SDA project, I would like to go for the "Burden of Proof" option. I'll select four experts in pharmacogenomics, personalized medicine, or bioethics: two who support implementation of this technology and two who go against it. I think this format would allow me to present both sides of an extremely nuanced debate, giving a better understanding of both the limitations and hopes of pharmacogenomics.
Journal 11:
For my SDA project, I’ve chosen the “Burden of Proof” format and will be creating a written piece that explores the debate surrounding pharmacogenomics and personalized medicine. This format allows me to present in-depth perspectives from experts on both sides of the issue, providing readers with a comprehensive understanding of pharmacogenomics and its implications. A written piece offers the advantage of detail and structure, enabling me to break down complex scientific information while making it accessible. By presenting arguments in an organized, reader-friendly way, I can offer an unbiased view of the potential benefits and challenges of pharmacogenomics, encouraging readers to consider the evidence and form their own opinions.
The structure of the written piece will begin with an introduction to pharmacogenomics, followed by a “pro” section highlighting how it aims to reduce adverse drug reactions, improve drug efficacy, and lower healthcare costs. This part will include success stories where personalized medicine has positively impacted patient outcomes. The “con” section will then address the challenges, such as limited access to genetic testing, ethical concerns surrounding genetic data, and the need for broader clinical trials across diverse populations. Each section will cite scientific studies, real-world examples, and expert opinions, allowing readers to understand the debate from multiple angles.
Sources and whatnot:
Phillips et al., JAMA highlights the high rates of adverse drug reactions in conventional medicine, emphasizing the need for personalized treatments. It reports that adverse drug reactions are a significant cause of hospitalizations, which underscores the potential impact of pharmacogenomics in reducing these incidents. This will be central to the “pro” section, illustrating how aligning treatments with genetic profiles could improve patient safety. Phillips et al. also discuss the economic benefits of pharmacogenomics, explaining how effective treatments lead to fewer medical interventions and lower healthcare costs. Including this will help support the claim that personalized medicine could make healthcare more efficient and sustainable.
Johnson and Relling gives some examples of how pharmacogenomics has been successfully applied, such as using genetic markers to guide drug choices in cancer treatment. I’ll use this to show how personalized medicine is already benefiting patients, offering readers concrete evidence of its potential. Johnson and Relling also discuss the limitations, particularly in terms of accessibility and the high cost of genetic testing. Including this perspective is important for addressing the “con” side, as it highlights real challenges that need to be addressed to ensure pharmacogenomics benefits all populations equitably.
CDC’s Genomics and Precision Health Initiative offers a broader government perspective, demonstrating the support that pharmacogenomics is receiving at the national level. This source is useful for showing that the push for personalized medicine is supported by reputable organizations, lending credibility to the argument for further investment in the field. However, the CDC also addresses concerns about health disparities, pointing out that precision medicine could deepen inequities if access to genetic testing is limited. This will help me to explore ethical concerns and show the need for balanced solutions in expanding pharmacogenomics.
Journal 12/SDA Reflection 2:
Making this SDA was both a challenging and rewarding experience. I dedicated about 10 hours in total, spread over several days, to complete the research, outline, and final draft. Much of this time was spent finding academic articles and trusted medical sources to ensure my arguments were supported by credible evidence. Writing the piece required both a deep understanding of pharmacogenomics and an ability to communicate complex ideas in an engaging and accessible way.
One of the key discoveries I made during this process was the sheer complexity and potential of pharmacogenomics. It was eye-opening to see how many lives could be improved or even saved with personalized medicine. At the same time, I was struck by how systemic issues—like unequal access to healthcare and insufficient research diversity—could prevent such a promising field from reaching its full potential. On a personal level, I realized how passionate I am about equity in healthcare and how much I value solutions that prioritize inclusivity.
The most important thing I learned was the balance between optimism and realism in addressing scientific advancements. While pharmacogenomics offers incredible possibilities, we must also critically examine its challenges to create a more equitable system. Looking back, I am proud of how I synthesized a complex topic into a cohesive narrative and incorporated direct quotes to support my analysis.
However, I recognize there’s room for improvement in terms of pacing and refining my argument. Some sections could have delved deeper into the ethical dimensions, and I want to work on integrating even more diverse perspectives. The knowledge I gained will undoubtedly inform future projects and discussions, especially as I pursue a career in bioengineering and personalized medicine.
My new essential question is: How can pharmacogenomic testing improve drug safety and efficacy for underrepresented populations in clinical research?
The main message of the podcast I selected emphasized the importance of environmental sustainability in urban planning. The creators likely chose this topic to highlight how everyday decisions can contribute to larger environmental goals. The podcast’s tone was informative yet conversational, making complex ideas approachable. The host employed storytelling techniques like brief anecdotes and statistics to ground the topic in real-world relevance. The sound design, including a calm narration and soft background music, helped maintain my focus and engagement.
This concise and engaging format inspired me to think about how I might use similar techniques in future presentations or written work, particularly in communicating science to non-specialist audiences.
Journal 13:
My partner and I will explore the development of tailored medicines for astronauts. This topic allows us to connect medicine, genetics, and epigenetics of astronauts, addressing a critical need for personalized healthcare in such critical environments. One key article for our research is "Translating Current Biomedical Therapies for Long-Duration, Deep Space Missions," published in Precision Clinical Medicine by Sonia Iosim, Matthew MacKay, Craig Westover, and Christopher Mason, examines how precision medicine and cellular engineering could mitigate health risks for astronauts. The authors analyze spaceflight hazards such as radiation, gravity, and isolation, and propose tailored pharmaceutical interventions, cellular therapies and genetic engineering as solutions to these challenges.
The article was accessed through PubMed using terms like "pharmacogenomics in astronomy," "precision medicine for astronauts," and "spaceflight health." The article reveals that astronauts face multiple risks, including "radiation-induced DNA damage, immune disfunction, and muscle atrophy due to microgravity." These physiological changes are exacerbated during long missions to destinations like Mars. It emphasizes the role of pharmacogenomics in tailoring drug regimens to individual genetic profiles to optimize safety and efficacy. For instance, the all-but-ignored enzyme CYP450, which metabolize many drugs, exhibit altered activity in space, as most enzymes do due to the epigenetic changes in the respective gene that codes for them. Additionally, the study highlights the potential for cellular engineering to enhance astronauts' resilience against radiation by leveraging proteins like Dsup, derived from tardigrades.
This study is vital to the field of space medicine and biophysics, as it shows the need for advanced biomedical strategies to support human exploration beyond Earth. By focusing on tailored solutions like cellular engineering and pharmacogenomics, it makes way for longer and safer missions. This approach could also help revolutionize healthcare on Earth by fostering innovations applicable to broader populations. Key terms such as "multi-omics," "cytokine profiling," and "Dsup protein" were new to me. Researching these terms broadened my understanding of both molecular biology and environmental stressors in space.
Listening to Radiolab's Animal Minds podcast offered insights into how emotion enhances storytelling. The narrative centered on a whale rescue, evoking empathy and curisoty while delivering scientific information. Inspired by this approach, we hope to make a compelling narrative that plays both into the science of pharmacogenomics with the human stories of astronauts.
Citation:
Iosim, Sonia, et al. "Translating Current Biomedical Therapies for Long-Duration, Deep Space Missions." Precision Clinical Medicine, vol. 2, no. 4, 2019, pp. 259-269. Oxford University press, doi:10.1093/pcmedi/pbz022
Journal 14:
The article “The Future of Personalized Medicine in Space: From Observations to Countermeasures” delves into the pressing need for personalized medicine to safeguard astronaut health and enhance mission outcomes. Space missions expose astronauts to extreme and unique environmental conditions, including microgravity, high radiation levels, and confinement, all of which impose significant physiological and psychological stresses. The primary problem addressed in the article is the interindividual variability in physiological responses to these stressors and the efficacy of pharmacological treatments in space. Current medical approaches often rely on standardized countermeasures, which fail to account for individual differences in genetics, immune responses, and other physiological traits. The purpose of the article is to propose strategies for implementing tailored medical solutions, from pre-mission genetic screening to individualized in-flight pharmacological interventions, to mitigate health risks and improve outcomes for astronauts.
Published in Frontiers in Bioengineering and Biotechnology, this article aligns with the journal’s goal of promoting interdisciplinary research to address challenges at the intersection of biology, engineering, and technology. The journal emphasizes solutions that have practical applications, such as advancements in personalized medicine, which not only benefit astronauts but also have potential implications for terrestrial healthcare.
The article is authored by a multidisciplinary team of experts, with the corresponding author, Elizabeth Pavez Loriè, leading the initiative. Loriè is affiliated with the Leibniz Institute for Environmental Medicine in Düsseldorf, Germany, where she focuses on environmental and space medicine. Another notable contributor is Virginia Wotring, a professor at the International Space University, whose research specializes in pharmacology for space missions and the physiological impacts of microgravity. These authors have significantly advanced the fields of space medicine and pharmacology by integrating genetic and physiological insights to address the unique challenges of human spaceflight.
This article builds upon and distinguishes itself from previous research by presenting a cohesive framework that integrates pharmacogenomics with space physiology. While last week’s article focused on general pharmacogenomics or the effects of space on human health, this study uniquely emphasizes how genetic and physiological variability can be leveraged to develop individualized healthcare strategies. It explores the use of pre-mission pharmacogenetic testing, in vitro modeling, and real-time health monitoring to create a holistic approach to astronaut care, bridging the gap between research on genetic variability and its practical application in space.
In collaboration with my partner’s research, this article forms a robust foundation for exploring tailored medicine for astronauts. My focus on pharmacogenomics integrates seamlessly with her study of space’s physiological effects, as this article highlights how genetic variability influences medication efficacy and physiological adaptation in space. For example, astronauts experience significant interindividual variability in drug metabolism and immune response, necessitating tailored pharmacological countermeasures to ensure mission success. This integrative approach supports the broader theme of combining genetic and environmental factors to optimize astronaut health.
The article provides several key data points relevant to my research question. For instance, it notes that 83% of astronauts experience medical events during missions, with notable variability in symptoms such as skin rashes and immune dysregulation. Additionally, preemptive pharmacogenetic testing is identified as a critical tool for improving medication efficacy and avoiding adverse reactions. The variability in drug absorption, distribution, and metabolism in microgravity further underscores the need for individualized medical strategies.
Despite its strengths, the article has some limitations. One major challenge is the small sample size of astronaut studies, which limits the generalizability of findings. Additionally, most pharmacokinetic data are derived from salivary rather than blood samples, reducing the precision of results. There is also a lack of longitudinal studies to fully understand the long-term effects of spaceflight on health and the efficacy of countermeasures. These gaps highlight the need for continued research to refine personalized medicine approaches for space.
The article introduced several new terms that enhance understanding of the topic. “Pharmacogenetics” refers to the study of how genetic variations influence drug responses, a core component of personalized medicine. “Microgravity” describes the condition of near weightlessness experienced in space, which significantly alters human physiology. Additionally, “endocannabinoids” were discussed as molecules that play a role in stress and immune regulation during space missions, illustrating the complex interplay between physiological systems in extreme environments.
To identify this article, I used the Frontiers in Bioengineering and Biotechnology journal database, searching terms such as “personalized medicine,” “space,” and “astronauts.” This strategic use of databases ensured access to high-quality, peer-reviewed research directly relevant to my topic.
Drawing inspiration from the Relative Genius podcast, the use of mystery and storytelling can effectively engage audiences when presenting complex scientific topics. The podcast maintained interest by building suspense around the search for Einstein’s brain, gradually revealing historical and scientific insights. Similarly, weaving a narrative around the challenges and breakthroughs in personalized astronaut medicine can make the subject more compelling and relatable. This approach can be particularly useful for communicating my findings and emphasizing the significance of tailoring healthcare solutions for space exploration.
Journal 15:
The article, Pharmacogenomics Guided Spaceflight: The Intersection Between Space-Flown Drugs and Space Genes, explores the challenges astronauts face with drug efficacy during spaceflight. The study identifies the problem of altered drug metabolism caused by microgravity and radiation, which significantly impact gene expression and biological pathways critical for drug absorption, metabolism, and excretion. The purpose of the research is to create a comprehensive database linking drugs commonly used during missions with genes affected by the space environment. This database aims to guide the development of precision medicine protocols for long-duration space exploration.
Published on bioRxiv, this preprint platform facilitates rapid access to new research, fostering global collaboration. The study’s open-access nature underscores the organizational goal of accelerating discovery in fields like aerospace medicine. The authors—Dr. Christopher Mason, Dr. Theodore Nelson, and Dr. Michael Schmidt—are leaders in genomics and space health. Mason, a genomics expert, has contributed extensively to NASA’s Twin Study, while Nelson specializes in molecular biology, and Schmidt focuses on human performance optimization in extreme environments. Collectively, their work significantly advances pharmacogenomics in space.
The study’s methods involved analyzing 218 drugs used in space missions and their interactions with 772 genes affected by space conditions. Researchers used multi-omics approaches, including genomics and proteomics, to predict gene-drug interactions under microgravity. Key findings revealed that genes in the CYP450 family, such as CYP2D6 and CYP3A4, show altered expression in space, affecting drug metabolism. For example, medications like statins and sleep aids exhibit reduced efficacy due to these genetic changes. However, the study relies on Earth-based data, which might not fully replicate the dynamic changes in astronaut physiology during spaceflight. Additionally, the small sample size of astronauts limits the generalizability of the results.
The findings have direct real-world applications, especially in developing personalized medicine for astronauts on missions to Mars or the Moon. This research aligns closely with the theme my partner and I are exploring: tailoring medicines for spaceflight. For instance, identifying polymorphisms in the SLCO1B1 gene could improve how astronauts metabolize statins, preventing adverse effects. The study highlights the potential of integrating pharmacogenomics into space health protocols, ensuring safety and performance in extreme environments.
The article introduced new key terms like multi-omics integration (combining genomics, transcriptomics, and proteomics for comprehensive biological analysis) and space genes (genes whose expression changes due to spaceflight). These terms provided deeper insights into the complexity of tailoring medicine for space environments. Higher-order thinking questions (HOTQs) raised by this research include: How can pharmacogenomics adapt to the dynamic physiological changes in space? and What role could AI play in predicting real-time gene-drug interactions during missions?
Listening to the Radiolab episode, The Queen of Dying, provided valuable insights into how storytelling and research intersect to create a compelling narrative. The story of Elisabeth Kübler-Ross’s work on understanding death was rooted in her personal observations and direct conversations with patients. Her persistence in uncovering truths about the dying process mirrored the scientific curiosity in our own research. For example, Kübler-Ross defied conventional norms by going room to room to find terminally ill patients, demonstrating how qualitative observation and patient interaction can reshape entire fields of study. Similarly, the research in pharmacogenomics relies on deep observation of genetic data and patient variability to innovate healthcare in space.
The storytelling in The Queen of Dying used vivid imagery, such as Kübler-Ross walking hospital hallways in Birkenstocks and aloha shirts, which humanized her groundbreaking work. This approach kept me engaged and made the science more relatable. It inspired me to think about how we can present pharmacogenomics in a way that resonates emotionally with our audience while highlighting its transformative potential.
Citation
Mason, Christopher E., et al. Pharmacogenomics Guided Spaceflight: The Intersection Between Space-Flown Drugs and Space Genes. bioRxiv, 2024. doi:10.1101/2024.01.16.575951.
Midterm Reflection
Collaboration was essential in creating our podcast, and my partner and I divided our responsibilities based on our strengths. While I focused on researching and structuring the script, my partner worked on refining our delivery and ensuring the flow of the conversation felt natural. We collaborated on the recording process, providing feedback to each other to improve our pacing and clarity. One of the biggest challenges we faced was aligning our schedules for recording and editing, but we overcame it by setting firm deadlines and communicating consistently.
Our creative vision evolved throughout the process. Initially, we thought we would simply present research in a structured format, but as we progressed, we realized the importance of storytelling and engaging transitions. We were surprised by how much adding sound effects and well-placed pauses enhanced the narrative. One creative breakthrough was realizing that framing our discussion as a thought experiment—placing the listener in an astronaut’s shoes—made the topic more immersive.
For research and preparation, we relied on peer-reviewed articles from bioRxiv, PubMed, and scholarly sources on pharmacogenomics and space medicine. Organizing our information was crucial, so we used shared documents to track key findings and citations. To stay on track, we established small milestones, such as completing the script before starting any recording.
Technically, we both developed new skills in scriptwriting, audio editing, and pacing our speech for clarity. The most challenging part was ensuring our delivery sounded natural while staying true to the scientific content. At first, our recordings felt too scripted, so we adjusted by rehearsing sections in a conversational style before recording the final takes.
The biggest obstacle we faced was balancing the depth of our research with keeping the podcast engaging. Some of the content was highly technical, and we had to simplify complex ideas without losing accuracy. We handled this by testing our explanations on others to see if they made sense to someone unfamiliar with the topic.
Through this project, I learned that effective communication is about more than just presenting information—it’s about crafting a narrative that keeps the audience engaged. I also gained a greater appreciation for teamwork and the importance of adaptability when working on a long-term project.
One of the proudest moments was hearing the final edited version of our podcast. The way the music, narration, and sound effects came together created a professional and compelling final product. It felt like all the hard work we put into refining our script and delivery had paid off.
If we had an extra week, we would improve the pacing of certain sections and potentially add an expert interview to enhance the credibility and depth of our discussion. Bringing in an outside perspective could have added a dynamic element to the podcast.
For future podcasters, my advice would be to focus on storytelling, not just facts. Engaging an audience requires making them care about the topic, and that means finding a hook, building a strong structure, and delivering the content with confidence. Also, don’t underestimate the importance of editing—it can transform a good podcast into a great one.
The skills from this project will be invaluable in future academic and professional settings. Whether it’s presenting research, conducting interviews, or explaining complex topics in an accessible way, the ability to synthesize and communicate information effectively is essential. This project has reinforced the importance of clear, engaging communication, and I know I’ll take these lessons with me into future scientific work.
Journal 16:
For my case study, I’ve chosen the story of Victoria “Tori” Gray, one of the first patients to receive CRISPR-based gene therapy for sickle cell disease. Her case is particularly compelling because it represents the real-world impact of gene editing on a patient who had been suffering from an inherited disorder with limited treatment options. Tori Gray, a woman from Mississippi, had battled severe pain crises and hospitalizations for most of her life due to sickle cell disease. In 2019, she became the first U.S. patient to undergo an experimental CRISPR gene-editing treatment at the Sarah Cannon Research Institute in Nashville. The treatment, known as exagamglogene autotemcel (exa-cel), was designed to reactivate fetal hemoglobin production, reducing the effects of sickled red blood cells.
What makes her case unique is the groundbreaking use of CRISPR-Cas9 technology in a real patient, marking a shift from theoretical gene therapy to tangible medical application. Just months after the procedure, Tori’s health improved dramatically, her pain crises stopped, and she was able to live without the constant fear of hospitalization. Her case provides a powerful example of how bioengineering and pharmacogenomics can transform lives by targeting the genetic root of disease.
In my documentary, I want to explore several key questions: How did CRISPR gene-editing work in Tori’s case, and what made it a viable treatment for sickle cell disease? What role did pharmacogenomics play in determining whether Tori was a good candidate for this therapy? What are the long-term implications of gene-editing treatments like CRISPR for patients with inherited disorders, and how accessible will these therapies be?
Two sources that solidify this case as legitimate are: Frangoul, Haydar, et al. “CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia.” New England Journal of Medicine, vol. 384, no. 3, 2021, pp. 252-260. This study documents the clinical trial that Tori participated in and provides scientific evidence of the therapy’s effectiveness. Doudna, Jennifer A. A Crack in Creation: Gene Editing and the Unthinkable Power to Control Evolution. Houghton Mifflin Harcourt, 2017. This book, co-authored by one of the pioneers of CRISPR, explains the science behind gene editing and its potential applications in medicine, including sickle cell disease.
Journal 17:
One of the biggest unanswered questions surrounding Tori Gray’s case and the use of CRISPR for sickle cell disease is the long-term safety and durability of gene-editing treatments. While early results have been promising, with patients like Tori experiencing a dramatic reduction in symptoms, we still don’t know how these genetic modifications will hold up over decades. Will the edited stem cells continue to produce healthy red blood cells for the rest of a patient’s life, or will additional treatments be needed? Another concern is off-target effects, where CRISPR may accidentally edit unintended parts of the genome, potentially leading to unforeseen health risks, such as cancer.
There are multiple perspectives on this case. From a scientific standpoint, researchers see CRISPR-based treatments as a revolutionary step in treating genetic diseases at their source rather than just managing symptoms. Many believe it could eventually become the standard of care for inherited disorders. From an ethical standpoint, concerns arise about the accessibility and cost of these treatments. Exagamglogene autotemcel (exa-cel), the CRISPR therapy used in Tori’s case, is projected to be one of the most expensive treatments in history. If only wealthy patients or those with the best insurance can afford it, will this technology widen existing healthcare inequalities? On a personal level, Tori Gray and other patients see this as a second chance at life. She went from enduring crippling pain crises to living symptom-free, a transformation that makes her an advocate for the therapy despite the uncertainties.
One surprising detail I’ve uncovered is that Tori’s own stem cells were removed, edited, and then reinfused into her body after undergoing high-dose chemotherapy. Unlike a traditional medication, this isn’t a simple injection—it’s an intensive medical process that requires hospital stays, similar to a bone marrow transplant. This underscores the complexity of gene-editing treatments and why they are not yet widely accessible.
If I could interview one expert related to this case, I would choose Dr. Haydar Frangoul, the hematologist who led the clinical trial that treated Tori Gray. He is a widely respected figure in gene therapy research and has authored multiple studies on CRISPR-based treatments for blood disorders. I would ask him: Based on current data, how confident are you that CRISPR will provide a lifelong cure for sickle cell disease? What are the biggest challenges in making this treatment widely available? How do you respond to ethical concerns about the cost and accessibility of gene-editing therapies?
Dr. Frangoul’s credibility stems from his leadership in clinical trials and his firsthand experience with patients undergoing CRISPR treatments. Speaking with him would provide crucial insights into both the science and the real-world applications of gene therapy.
Journal 18
The documentary will begin with a compelling introduction that immediately draws viewers into Tori Gray’s story. It will open with archival footage or personal clips of Tori before and after her treatment, showing the stark contrast between her life with sickle cell disease and the transformation she experienced afterward. The narration will pose a thought-provoking question: “For decades, sickle cell disease was a life sentence of pain, hospitalizations, and limited treatment options. But what if the script could be rewritten—at the genetic level?” This will establish the stakes of the documentary while introducing CRISPR as a revolutionary tool that could fundamentally change the future of medicine. Background music with futuristic and emotional tones will set the mood, reinforcing the idea that science is at the threshold of a groundbreaking medical breakthrough.
Following the introduction, the documentary will shift into a background section that provides essential context for viewers unfamiliar with sickle cell disease and gene therapy. Using engaging 3D animations, I will illustrate how sickled red blood cells block blood flow, leading to excruciating pain crises and long-term organ damage. This section will also explore the history of sickle cell treatments, highlighting how past therapies like hydroxyurea and bone marrow transplants have had limited success due to accessibility issues and the need for a genetic match. At this point, I will introduce CRISPR-Cas9 through an animation showing how gene editing works at the molecular level, emphasizing its potential to correct genetic errors at their source. Expert interviews will provide additional insight, explaining why gene therapy represents a paradigm shift in treating inherited diseases.
The heart of the documentary will focus on Tori Gray’s case, following her journey from years of debilitating pain to becoming one of the first U.S. patients to undergo CRISPR-based therapy for sickle cell disease. This section will feature interview clips of Tori describing her struggles before treatment—frequent hospitalizations, severe pain, and the uncertainty of living with a life-threatening condition. I will also showcase footage of her undergoing the treatment process at the Sarah Cannon Research Institute, highlighting key moments such as the extraction of her stem cells, the CRISPR gene-editing procedure, and the reinfusion process after high-dose chemotherapy. Researchers and doctors involved in her case will provide commentary, explaining how the treatment works and why her case is a turning point in precision medicine. To make the science accessible, I will use split-screen visuals comparing Tori’s original blood cells to the genetically edited ones, showing how CRISPR successfully reactivated fetal hemoglobin production to counteract the effects of sickle cell disease.
The analysis section will dive deeper into the broader implications of CRISPR-based gene therapy, breaking it down into three key perspectives: scientific, ethical, and medical. From a scientific standpoint, I will explore how pharmacogenomics played a role in ensuring Tori was a good candidate for the treatment and how researchers determined that reactivating fetal hemoglobin was a viable strategy. From an ethical perspective, I will address pressing concerns surrounding gene-editing treatments, including their high costs, limited accessibility, and the potential for genetic inequality if only the wealthy can afford them. Finally, from a medical standpoint, I will discuss the long-term effects of gene editing—whether CRISPR-modified cells will remain stable for life and what risks, such as off-target effects, still need to be addressed.
The documentary will conclude by looking toward the future of gene therapy. What other diseases could CRISPR potentially cure? What barriers—scientific, financial, or ethical—must be overcome before these treatments become widely available?
Journal 19
The biggest takeaway I want my audience to remember is that gene therapy and pharmacogenomics have the potential to revolutionize medicine, but their accessibility, ethical implications, and long-term effects must be carefully considered. Tori Gray’s case is proof that CRISPR can successfully alter human DNA to treat genetic disorders, but it also raises critical questions: Who gets access to these treatments? Are they safe in the long run? And what does this mean for the future of human genetic modification? By telling this story, I hope to inspire my audience to think deeply about both the promises and challenges of gene therapy.
Researching this case has given me a much deeper appreciation for the complexity of pharmacogenomics and bioengineering. Initially, I saw gene therapy as a straightforward fix—correct a mutation, and the disease is gone. But through studying CRISPR and cases like Tori Gray’s, I’ve realized that it’s not that simple. Even with a successful edit, there are long-term effects to consider, such as potential off-target mutations or unforeseen immune responses. Furthermore, I’ve gained a greater awareness of the ethical and socioeconomic issues surrounding gene therapy. The cost of treatment—estimated at nearly $2 million per patient—raises serious concerns about medical equity. Will this technology remain a luxury for the wealthy, or can it be scaled to benefit a broader population? These questions have reshaped how I view the future of medicine and the responsibility that comes with genetic engineering.
Before filming, I need to gather a few more sources to strengthen my documentary’s credibility and provide a well-rounded perspective:
An interview or statement from a bioethicist discussing the long-term implications of gene editing and the potential risks.
A scientific paper or study on the long-term stability of CRISPR-modified cells, addressing whether these edits remain effective for a lifetime or if additional treatments are needed.
A historical perspective on sickle cell disease treatment to highlight how far medicine has come and why CRISPR is such a breakthrough.
Patient testimonials beyond Tori Gray’s case to see if similar results have been achieved with other individuals.
Storyboard & Production Plan:
Pre-Production (Planning & Research)
Gather all necessary research materials and finalize sources.
Write the full script, breaking it into sections: Introduction, Background, Case Study, Analysis, and Conclusion.
Outline specific interview questions if expert commentary is included.
Create a visual storyboard to map out key scenes, including animations, archival footage, and interviews.
Filming & Production
Introduction Scene:
Footage of Tori Gray pre- and post-treatment to highlight her transformation.
Voiceover introducing CRISPR as a revolutionary technology.
Background music with an emotional yet futuristic tone.
Background Section:
3D animation showing how sickle cell disease affects red blood cells.
Archival images of past sickle cell treatments, such as early bone marrow transplants.
CRISPR animation demonstrating how the genetic edit is made.
Case Study – Tori Gray’s Journey:
Footage of her describing her experience with sickle cell before the treatment.
Clips from the Sarah Cannon Research Institute showing the treatment process.
Interviews or voiceovers from scientists explaining how CRISPR corrected her mutation.
Analysis & Ethical Discussion:
Interview with a bioethicist discussing access to gene therapy.
Breakdown of treatment costs and accessibility concerns.
Expert commentary on long-term risks and potential applications beyond sickle cell.
Conclusion:
Final thoughts from Tori on her new quality of life.
A montage of expert opinions on the future of gene therapy.
Closing question: “Is gene therapy the medical miracle of our time, or just the first step in an even greater ethical debate?”
Post-Production (Editing & Finalizing)
Editing Stage 1: Assemble all footage and organize scenes in sequence.
Editing Stage 2: Add voiceovers, expert interviews, and background music.
Editing Stage 3: Insert animations and on-screen text to explain scientific concepts.
Final Review: Watch the full documentary, make final adjustments, and add subtitles for accessibility.