1st Place Essay
Beyond the Genome: How Genetic Diversity Shapes the Future of Global Health
Snigdha Rai Jamnabai Narsee International School, Mumbai, India
Beyond the Genome: How Genetic Diversity Shapes the Future of Global HealthWhy does one child survive malaria while another fails to recover, even with similar treatment? The answer may lie deep down in their DNA. Genetic diversity, the variation in DNA sequences among individuals and populations, shapes how humans respond to diseases, drugs, and environmental pressures. Despite advances in genome sequencing, much of our knowledge is based on a limited segment of the population. Populations in Africa, Asia, and Indigenous communities remain vastly underrepresented in genetic research.
References: • Gabai-Kapara, E., Lahad, A., Kaufman, B., et al. Population-based screening for breast and ovarian cancer risk due to BRCA1 and BRCA2. Nature Genetics, 2014. https://www.nature.com/articles/ng1096-185 • Petersen, G. M., & Parmigiani, G. Genetic testing for cancer susceptibility: Lessons from BRCA1/2. CA: A Cancer Journal for Clinicians, 2005. https://pubmed.ncbi.nlm.nih.gov/15715976/ • National Center for Biotechnology Information (NCBI) Understanding Population Stratification (Chapter in: Genetic Variation and Its Functional Consequences). NCBI Bookshelf, 2008. https://www.ncbi.nlm.nih.gov/books/NBK20363/ • Spratt, D. E., Chan, T., Waldron, L., et al. Racial/Ethnic Disparities in Genomic Sequencing. JAMA Oncology, 2016. https://pmc.ncbi.nlm.nih.gov/articles/PMC7359278/ • Popejoy, A. B., & Fullerton, S. M. Genomics is failing on diversity. Nature, 2016. https://pmc.ncbi.nlm.nih.gov/articles/PMC9904154/ • Sirugo, G., Williams, S. M., & Tishkoff, S. A. The missing diversity in human genetic studies. Cell, 2019. https://www.sciencedirect.com/science/article/pii/S0198885917305104 • Vassy, J. L., Christensen, K. D., Schonman, E. F., & Green, R. C. The impact of genotype disclosure on health behavior. Journal of General Internal Medicine, 2014. https://pmc.ncbi.nlm.nih.gov/articles/PMC4143101/ • Kurian, A. W., Hare, E. E., Mills, M. A., et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. JAMA Oncology, 2015. https://pmc.ncbi.nlm.nih.gov/articles/PMC9621418/ • Ramos, E. M., & Rotimi, C. N. Population representation in genomic studies: Implications for precision medicine. Frontiers in Genetics, 2023. https://pmc.ncbi.nlm.nih.gov/articles/PMC9733397/ • Fackenthal, J. D., & Olopade, O. I. Breast cancer risk associated with BRCA1 and BRCA2 in diverse populations. Oncogene, 2003. https://pubmed.ncbi.nlm.nih.gov/9863508/
2nd Place Essay
Symphony of Our Genes: A Journey into Genetic Diversity and Human Health
Mei Bejdo Gjimnazi Sami Frasheri, Tirane, Albania
Beneath our skin, beyond the mirror of our eyes, lies a hidden language—one not spoken, but written in four letters: A, T, C, and G. These letters, strung together in intricate sequences, compose the 20,000-or-so genes that orchestrate the very essence of who we are. But no two symphonies are alike. Just as each snowflake spirals uniquely from the clouds, each human genome holds subtle variations—some barely whispering their presence, others shouting across generations. This invisible mosaic of genetic diversity is not just a marvel of nature, but the compass guiding modern medicine. To understand this diversity is to understand why one person succumbs to disease while another resists, why a drug heals one and harms another. In a world racing toward personalized healthcare, genetic diversity is the map—and ignorance of it is a risk we can no longer afford.
Imagine two hearts, both beating in rhythm, yet one falters after surgery despite standard treatment. The culprit? A single gene named CYP2C19, a quiet engineer of metabolism. For many, this gene processes clopidogrel, a blood-thinner essential for preventing clots. But for others—those carrying the CYP2C19 *2 or *3 variants—this gene malfunctions, leaving the medication inert. The result? A heart unprotected, a life endangered. In such moments, the beauty of genetic testing unfolds—not as a distant promise, but as an urgent necessity. This is the frontier where knowledge of DNA saves lives. Doctors informed of a patient’s genotype can pivot to safer, more effective treatments, bypassing the peril of a one-size-fits-all prescription. It is here, in these precise pivots, that genetic diversity transforms from abstract data into lifesaving action.
But genes do more than shape our response to drugs—they inscribe the stories of our vulnerabilities and strengths. Enter BRCA1 and BRCA2, names etched in medical history. Variants in these genes, found disproportionately in some populations, drastically elevate the risk of breast and ovarian cancers. Yet, knowledge is power. Women who discover these mutations through genetic screening are no longer bound to fate; they’re armed with options—from enhanced monitoring to preventative surgery. In such cases, understanding genetic diversity becomes a shield, crafted not of metal, but of molecular foresight. It empowers individuals to rewrite their destinies before illness writes them instead.
Zooming out from individual genes, genetic diversity also offers a lens into population-level resilience and risk. The HBB gene, mutated in sickle cell disease, is one such paradox. Carriers of the sickle trait, predominantly from African and Mediterranean ancestry, inherit a defense against malaria—a deadly foe in their ancestral lands. Yet this evolutionary shield comes with cost: a risk of sickle cell complications. This genetic give-and-take reminds us that diversity is not just biological but historical, woven through centuries of migration, adaptation, and survival. Any health system blind to such context is doomed to misdiagnose, mistreat, and misunderstand the very people it seeks to heal.
As we peer into the genome’s glowing code with the tools of modern science, one truth becomes clear: understanding genetic diversity is not optional; it is foundational. It is the path to equitable medicine, to a future where treatment is tailored not by guesswork, but by genetic insight. Initiatives like the 1000 Genomes Project and the European Society of Human Genetics’ outreach through EuroGEMS are already lighting this path. But we must walk it faster. We must fund research, educate clinicians, and engage communities. Only then can we transform our collective genetic symphony—from a scattered hum into a chorus of healing.
References: Scott, S. A., et al. (2013). Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C19 and Clopidogrel Therapy. Clinical Pharmacology & Therapeutics. Mavaddat, N., et al. (2013). Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers. Cancer Epidemiology, Biomarkers & Prevention Tishkoff, S. A., et al. (2001). Haplotype diversity and linkage disequilibrium at human G6PD: Recent origin of alleles that confer malarial resistance. Science.
3rd Place Essay
Understanding Genetic Diversity
Basak Memiguven Cankaya Doga Anadolu Lisesi, Ankara, Turkey
Why Understanding Genetic Diversity Is Important for Health and DiseaseEach person is different, and a big reason for that lies in our genes. Genes are made of DNA, and small variations in DNA from one person to another—known as genetic diversity—can affect our health in many ways. These differences help explain why some people are more likely to develop certain diseases, why medications work differently for different people, and why some inherited conditions are more common in certain groups.Genetic diversity is a natural result of human evolution. Over thousands of years, people in different parts of the world developed slightly different versions of genes due to changes in their environments, diets, and lifestyles. These genetic changes, passed down through generations, can affect health today in both helpful and harmful ways. Understanding these differences is becoming more essential in medicine. With more knowledge about how genes influence our health, doctors can better predict, prevent, and treat diseases based on a person’s unique genetic profile.A clear example is how individuals respond to medications. The CYP2D6 gene helps the body break down a wide range of common drugs, including pain relievers and antidepressants. But not everyone has the same version of this gene. Some people metabolize drugs more slowly, while others break them down too quickly. For instance, the CYP2D6 4 variant works less efficiently and is more common in Europeans. In contrast, people from North Africa or the Middle East may carry extra copies of the gene, causing faster drug metabolism. This means a standard dose might be too strong or too weak depending on the person’s genes.Another important gene is BRCA1, which is linked to higher risks of breast and ovarian cancers when it has certain mutations. One such mutation, called 185delAG, appears more often in Ashkenazi Jewish populations. If someone is known to carry this mutation, doctors can recommend more frequent screenings or even preventive treatments. This shows how learning about specific genetic differences can lead to life-saving decisions.Genes also play a role in how our bodies handle infections. The CCR5 gene, for example, makes a protein that HIV uses to infect cells. A version of the gene called CCR5-Δ32 changes the protein, offering protection against HIV. People with two copies of this variant are very resistant to HIV, while those with one copy may experience slower disease progression. This variant is most common in Europeans and rare in other groups. Discovering this has helped researchers explore new ways to prevent and treat HIV.Another gene, APOE, especially the APOE ε4 variant, is linked to a higher chance of developing Alzheimer’s disease. But the risk level depends on a person’s background. For example, African Americans with this gene may have a lower risk than Europeans with the same variant. This shows how ancestry and overall genetic background can influence how a gene affects health.Genetic diversity also explains simpler traits. For instance, lactose intolerance is more common in people of East Asian, African, and Native American ancestry, while many Europeans can digest milk into adulthood. This is due to a variation near the LCT gene, which controls the enzyme that breaks down lactose. Knowing this helps provide better nutrition advice based on genetic background.Understanding genetic diversity is also important for fairness in healthcare. In the past, most genetic studies focused on people of European descent. As a result, tests and treatments often don’t work as well for other populations. For example, sickle cell disease is caused by a mutation in the HBB gene, which is more common in people of African, Middle Eastern, or Mediterranean descent. If doctors don’t consider a patient’s background, they may miss or misdiagnose this condition.Fortunately, large studies like the 1000 Genomes Project and the All of Us Research Program are now collecting genetic data from diverse populations. These efforts aim to improve our understanding of genetic diversity across all groups, making healthcare more inclusive and accurate for everyone.In summary, genetic diversity plays a vital role in how diseases appear, how people respond to medications, and how treatments are designed. Differences in genes like CYP2D6, BRCA1, CCR5, and APOE help explain why health outcomes vary between individuals. Even everyday health traits, like lactose digestion, are affected by our genes. By studying and respecting genetic differences, doctors and scientists can offer more precise and fair medical care. To make healthcare truly effective, we must include people of all backgrounds in genetic research.
References:
Liu, L., et al. (2017). The role of CYP2D6 in the metabolism of drugs. Pharmacogenomics Journal.Easton, D. F., et al. (2015). Genome-wide association study identifies multiple loci for breast cancer risk. Nature Genetics.Samson, M., et al. (1996). Resistance to HIV-1 infection due to a mutation in the CCR5 gene. Nature.Corder, E. H., et al. (1993). Protective effect of apolipoprotein E type 2 allele for Alzheimer’s disease. Nature.Bersaglieri, T., et al. (2004). Genetic analysis of lactase persistence in European populations. Human Genetics.Grosse, S. D., et al. (2011). The economics of sickle cell disease: An overview. American Journal of Preventive Medicine.Churchill, R., et al. (2018). Inclusion of diverse populations in genetic research. The Lancet.
Essays Honourable Mention 1
The Importance of Understanding Genetic Diversity in Health and Disease
Tuna Kurtoglu ÖZEL SIVAS MODERN BILIMLER AKADEMISI, Sivas, Turkey
Over the past century, geneticists have focused on identifying genetic traits associated with specific populations, such as lactose intolerance and sickle cell anemia. These efforts often targeted isolated groups, enabling the discovery of certain variants with strong health implications. However, as genomic technologies continue to evolve, there is a growing need to broaden the scope of research to include more diverse global populations. Understanding genetic diversity is essential not only for scientific completeness but also for ensuring equity in healthcare outcomes. Recent advancements in genomic research have led to the creation of large biobanks such as the UK Biobank, the Estonian Biobank, and the FinnGen project. These initiatives collect genomic and health data from thousands of individuals, allowing researchers to uncover links between genes and diseases. While these projects have greatly expanded our understanding of human genetics, they are still limited in terms of population diversity. Most of the data comes from individuals of European descent, leaving gaps in our knowledge of genetic variations in other ethnic groups. This lack of diversity can have serious consequences. For example, a variant in the HBB gene causes sickle cell anemia, a disease prevalent among individuals of African ancestry. If genetic studies are predominantly based on European populations, such variants may be overlooked, leading to misdiagnosis or inadequate treatment options for affected individuals. Similarly, the ALDH2 gene, which affects alcohol metabolism, is commonly found in East Asian populations but is rare elsewhere. Failure to account for such population-specific variants may result in inaccurate assessments of disease risk or drug response. Genetic diversity also plays a critical role in pharmacogenomics—the study of how genes affect a person’s response to drugs. One well-documented example is the gene CYP2D6, which metabolizes many commonly prescribed medications, including antidepressants and opioids. Variants in CYP2D6 can categorize individuals as poor, intermediate, extensive, or ultra-rapid metabolizers. These differences can lead to adverse drug reactions or therapeutic failure if not taken into account. Importantly, the frequency of these variants varies across populations, highlighting the need for inclusive research. Another example involves the PCSK9 gene, which regulates cholesterol levels. Variants in this gene have led to the development of new cholesterol-lowering drugs known as PCSK9 inhibitors. Interestingly, the most impactful variants were first identified in African American populations, who have a higher frequency of PCSK9 loss-of-function mutations. This discovery underscores the value of studying diverse groups, not only for understanding disease but also for driving innovation in treatment. Moreover, failing to include diverse populations can lead to biased polygenic risk scores (PRS), which estimate an individual’s genetic risk of developing certain diseases. These scores are typically derived from genome-wide association studies (GWAS), which rely on large sample sizes. However, when the majority of participants are of European ancestry, PRS accuracy diminishes when applied to other populations. This limits the clinical utility of genetic testing for underrepresented groups and may exacerbate existing health disparities. To address this issue, researchers must actively recruit participants from a broad range of backgrounds and ensure that their data is adequately represented in genetic studies. Efforts such as the All of Us Research Program in the United States aim to collect genetic information from one million diverse participants. Similarly, initiatives like H3Africa work to build genomics research capacity on the African continent, promoting the inclusion of African populations in global genetic databases. In conclusion, understanding genetic diversity is not just a scientific imperative but also a matter of social justice. By including diverse populations in genomic research, we can improve the accuracy of genetic testing, enhance our understanding of disease mechanisms, and develop more effective, personalized treatments. As we move forward in the genomic era, it is essential that no population is left behind.
References: 1. Bycroft, C., et al. (2018). The UK Biobank resource with deep phenotyping and genomic data. Nature, 562(7726), 203-209. 2. Leitsalu, L., et al. (2015). Cohort profile: Estonian Biobank of the Estonian Genome Center, University of Tartu. International Journal of Epidemiology, 44(4), 1137-1147. 3. Kurki, M.I., et al. (2023). FinnGen: Unique genetic insights from combining isolated population and national health register data. Nature, 616(7957), 775-782. 4. Bentley, A.R., Callier, S.L., & Rotimi, C.N. (2020). Diversity and inclusion in genomic research: Why the uneven progress? Journal of Community Genetics, 11(3), 319-326. 5. Manrai, A.K., et al. (2016). Genetic misdiagnoses and the potential for health disparities. New England Journal of Medicine, 375(7), 655-665.
Essays Honourable Mention 2
How Genetic Diversity Helps Us Understand Disease
Sarp Sabancilar ÇANKAYA DOGA 86;A ANADOLU LİSESİ, Ankara, Turkey
Every person is different. We have different eye colors, hair types, body shapes, and even how we respond to medicines or get sick. These differences come from our DNA. The small changes in our DNA from person to person are called genetic diversity. This diversity is very important in understanding health and disease. By studying genetic diversity, scientists can find out why some people get certain illnesses, why some medicines work better for some than others, and how we can make healthcare more fair for everyone. DevelopmentIn the past, scientists often studied small, isolated populations. These studies led to important discoveries. For example, people with African ancestry often carry a version of the HBB gene that protects them from malaria. However, this same gene variant can also cause sickle cell disease, a serious blood condition [Piel et al., Lancet, 2017]. Another example is lactose intolerance. In most parts of the world, adults cannot digest milk. But in Northern Europe, many people can, thanks to a gene variant near the LCT gene [Ingram et al., Human Genetics, 2009]. Today, big genetic projects like the UK Biobank, FinnGen, and Estonian Biobank help scientists study the DNA of millions of people. These projects have helped discover genes linked to diseases like cancer, diabetes, and heart disease. But there is a problem: most of these studies are done on people of European ancestry [Sirugo et al., Cell, 2019]. This means we are missing important information from other populations, like those in Africa, Asia, or Latin America. This lack of diversity can cause problems in healthcare. A good example is the APOL1 gene. Many people with African ancestry have a version of this gene that protects them from African sleeping sickness. But the same version also increases the risk of kidney disease [Genovese et al., Science, 2010]. If doctors do not know about this, they might not understand why kidney problems are more common in certain groups. Another gene, HLA-B*57:01, is more common in Europeans and can cause a serious reaction to a medicine called abacavir, used to treat HIV. Doctors test for this gene before giving the medicine [Mallal et al., NEJM, 2008]. But in other populations, this gene is rare. Without knowing the genetic differences, testing might not always be necessary or helpful. Scientists also use polygenic risk scores (PRS) to predict how likely someone is to get a disease. But since these scores are mostly based on European DNA, they are not always accurate for people from other backgrounds [Martin et al., Nature Genetics, 2019]. This can lead to unfair or wrong results in healthcare. To solve this problem, researchers are working on collecting more diverse genetic data. Projects like H3Africa, which focuses on African genetic studies, and All of Us, a U.S. project that collects DNA from many different groups, help include people from different regions and ethnicities. This is important to make sure medical research helps everyone equally. ConclusionGenetic diversity is not just about where we come from — it’s about how we stay healthy and how medicine works for us. By understanding the genetic differences between people, scientists can find better ways to prevent, diagnose, and treat diseases. Examples like the APOL1 and HLA-B57:01* genes show us how important it is to include all populations in genetic research. Without diversity, we risk creating a healthcare system that only works for some people. With diversity, we can build a future where medicine is personalized, fair, and effective for everyone.
References: 1:Sirugo, G., Williams, S.M., & Tishkoff, S.A. (2019). The Missing Diversity in Human Genetic Studies. Cell. 2:Genovese, G., et al. (2010). Association of Trypanolytic ApoL1 Variants with Kidney Disease in African Americans. Science. 3:Mallal, S., et al. (2008). HLA-B5701 Screening for Hypersensitivity to Abacavir*. New England Journal of Medicine. 4:Martin, A.R., et al. (2019). Clinical use of current polygenic risk scores may exacerbate health disparities. Nature Genetics. 5:Ingram, C.J.E., et al. (2009). Lactose digestion and the evolutionary genetics of lactase persistence. Human Genetics. 6:Piel, F.B., et al. (2017). Sickle cell disease. Lancet.
Essays Honourable Mention 3
Genetic Diversity : The Depth of One Percent
Minseo Kim Carmel High School, United States
More than 99% of the human genome is shared, meaning that only around 1% or less of your genetic material is what makes you different from other people[1]. Despite such a quite literal molecular difference, that genetic variety is what makes the human system and healthcare so difficult to generalize. Difference in genetic makeup creates different responses to medicine, different metabolic characteristics, and even different makeup of common diseases in a population. Throughout history, the importance of genetic diversity had been underestimated and clinical studies were often inaccessible or disregarding social minority populations. Such negligence and the lack of data led to severe progression of disease and even fatal results. In the present, scientists and healthcare workers are studying and archiving a variety of genes to better understand the diverse expression of the human body and to create management and prevention standards that can help reduce risks and healthcare costs for many. However, some populations still remain underrepresented, signifying that the science community still has to work towards the formation of a more diverse and inclusive research environment. Genetic difference calls for different medical procedures and medicine dosages. A 2006 study reports that individuals with variants of the RYR1 gene have a greater risk of Malignant Hyperthermia, a condition where certain volatile anesthetics can cause fatal symptoms such as fever, abnormal muscle contraction, and tachycardia[2]. Patients with this genetic variant could suffer critical harm if they or their healthcare organizations were not knowledgeable of their condition. The dosage and efficacy of medical drugs is also different among individuals with different genetic makeup. CYP2C19 is a liver enzyme that catalyzes the breakdown and metabolism of drugs. Variants in this gene result in different manner, efficiency, and rate of drug absorption[3]. A 2019 study on Clopidogrel, an antiplatelet medication, reports that individuals with two copies of non-functional alleles of CYP2C19 have reduced activation rate of Clopidogrel, resulting in a higher chance of cardiovascular event compared to other patients with functional alleles[4]. A one-fits-all medical standard would put patients at constant risk of being poorly treated with widely generalized medications that have little or adverse effect to them. Being familiar and well-informed of genetic variants is crucial to long-term management of diseases as well. Sickle Cell Disease is an inherited disease that causes red blood cells to polymerize into crescent shapes under oxidative stress due to a point mutation in the HBB gene[5]. Patients of Sickle Cell Disease experience medical crises under situations demanding high respiration and metabolism. When the doctor provides their patient with health instructions, being aware of underlying genetic complications like Sickle Cell Disease that could impact the feasibility of a health plan can help the patient receive an effective, actionable goal that does not contradict their genetic disease management. Acknowledging and studying diverse genetic variants and their impact on human health can make the healthcare system trustable and accessible for more people with diverse conditions. Despite the importance of genetic diversity in healthcare, many variants of genes and the corresponding population still remain underexplored. Although decreasing in number, some studies continue to resort to an overrepresented study sample, giving hastily generalized conclusions that are inapplicable to a number of real-life patients. The fact that DNA is a highly personal component also contributes to the difficulty of trying to reflect real-life genetic diversity to the research environment; genetic material cannot be regularly taken in great amounts in numerous communities at random. Therefore, individual interest in one’s own genetic makeup and the willingness to scientifically confirm it is the most important factor for the science community to achieve high resemblance to the actual population in their experiment. To encourage research participation of underrepresented populations, the role genetics play in the shaping of human health should be taken seriously in healthcare services and widely educated to the public.
References: [1] Collins, Francis S., and Monique K. Mansoura. “The Human Genome Project.” Cancer, vol. 91, no. S1, 1 Jan. 2001, pp. 221-225, https://doi.org/10.1002/1097-0142(20010101)91:1+<221::aid-cncr8>3.0.co;2-9. [2] Robinson, Rachel, et al. “Mutations in RYR1 in malignant hyperthermia and central core disease.” Human Mutation, vol. 27, no. 10, Oct. 2006, pp. 977-989, https://doi.org/10.1002/humu.20356. [3] Qamar Shubbar, Aminah Alchakee, Khaled Walid Issa, Abdul Jabbar Adi, Ali Ibrahim Shorbagi, & Maha Saber-Ayad. (2024). From genes to drugs: CYP2C19 and pharmacogenetics in clinical practice. Frontiers in Pharmacology, 15. https://doi.org/10.3389/fphar.2024.1326776 [4] Pereira, N. L., Rihal, C. S., So, D. Y. F., Rosenberg, Y., Lennon, R. J., Mathew, V., Goodman, S. G., Weinshilboum, R. M., Wang, L., Baudhuin, L. M., Lerman, A., Hasan, A., Iturriaga, E., Fu, Y.-P., Geller, N., Bailey, K., & Farkouh, M. E. (2019). Clopidogrel Pharmacogenetics. Circulation: Cardiovascular Interventions, 12(4). https://doi.org/10.1161/circinterventions.119.007811 [5] Elendu, C., Amaechi, D. C., Alakwe-Ojimba, C. E., Elendu, T. C., Elendu, R. C., Ayabazu, C. P., Aina, T. O., Aborisade, O. B., & Adenikinju, J. S. (2023). Understanding Sickle Cell disease: Causes, symptoms, and Treatment Options. Medicine, 102(38), e35237-e35237. https://doi.org/10.1097/md.0000000000035237
Essays Honourable Mention 4
Every Genome Counts
Arwen Shah Greenwood High, Babgalore, India
Prescribing the same drug dosage to a Norwegian farmer, a Maasai pastoralist, and a Japanese office worker would defy both logic and biology—yet this is precisely the blind spot created by Eurocentric genomic research. The human genome varies more across populations than traditional medical studies acknowledge, with consequences ranging from ineffective treatments to undiagnosed diseases. While isolated populations like the Finns and Ashkenazi Jews have provided crucial insights through ‘founder effects’ [1], their genetic homogeneity cannot represent humanity’s complexity. Modern biobanks, though revolutionary, replicate this bias: the UK Biobank’s dataset is 94% European, and GWAS studies overrepresent Europeans by 80%. This exclusion has tangible costs[2]. Consider APOL1, where G1 and G2 variants—selected in West Africans for resistance to trypanosomiasis—increase kidney disease risk [3]. These alleles went unnoticed for decades because early renal studies focused on Europeans, delaying critical screening protocols for millions of African-descended patients. Pharmacogenomics reveals even starker disparities. Warfarin, a blood thinner, requires precise dosing guided by VKORC1 and CYP2C9 variants. Europeans with the VKORC1 rs9923231-T allele metabolize the drug slowly, needing lower doses, while many Africans lack this variant and require higher doses [4]. Early guidelines based on European data led to dangerous overdoses in African patients, increasing hemorrhage risk. Similarly, the antiplatelet drug clopidogrel depends on CYP2C19 for activation. East Asians frequently carry loss-of-function alleles (CYP2C19*2/*3), rendering the drug ineffective and elevating stroke risk—a problem overlooked until population-specific studies revealed it [5]. The painkiller codeine’s metabolism tells a parallel story: ultra-rapid CYP2D6 metabolizers, common in Ethiopia, convert codeine to morphine too efficiently, risking fatal overdose in infants through breast milk [6], while slow metabolizers in East Asia may get no pain relief at all. These examples underscore that precision medicine cannot be “precise” without diverse data. Disease susceptibility also follows population-specific patterns. The SLC16A11 variant, present in 50% of Native American-derived groups but rare in others, increases type 2 diabetes risk by disrupting lipid metabolism—a discovery made only when researchers included Mexican participants [7]. Similarly, PCSK9 variants like Y142X in African Americans and R46L in Europeans lower LDL cholesterol and reduce heart attack risk by up to 88%, inspiring blockbuster PCSK9 inhibitor drugs [8]. Yet these breakthroughs nearly missed populations excluded from early studies. Even protective mutations reflect evolutionary trade-offs: the CCR5-Δ32 deletion, found in 10% of Europeans, confers HIV resistance but increases West Nile virus susceptibility [9], while APOL1’s kidney risks accompany its parasite defense. Such nuances vanish when research ignores diversity. Convergent evolution further complicates the picture. Lactose tolerance, often mislabeled a “European” trait, arose independently in African pastoralists (via the -14010C mutation), Middle Easterners (-13915T), and Northern Europeans (-13910T) [10]. Each population evolved lactase persistence through distinct genetic pathways—a fact obscured when studies focus on one group. Similarly, sickle cell mutations protect against malaria not only in Africans but also in Greeks and Indians, yet Eurocentric narratives historically framed it as an “African disease” [11]. These oversimplifications hinder both scientific understanding and clinical care. The ethical and practical consequences are profound. Transthyretin (TTR) amyloidosis, a deadly cardiac condition, was misclassified as “rare” in Africans until H3Africa research identified it as a major cause of heart failure in Ghana and Kenya [12]. Similarly, 275 million genetic variants in the NIH’s All of Us Program—which prioritizes diversity—were absent from prior databases [13]. Without such inclusion, precision medicine risks exacerbating disparities: polygenic risk scores for breast cancer, developed using European data, fail to predict risk accurately in African American women, delaying lifesaving screenings. Initiatives like H3Africa and All of Us are correcting these gaps, but systemic change requires deeper shifts. Embedding pharmacogenomic testing into routine care, as Mayo Clinic’s RIGHT Study did for CYP2C19 and CYP2D6 [14], could prevent thousands of adverse drug reactions. Global biobanks, from Qatar Genome to Singapore’s SG10K, must become the norm, not exceptions. Most critically, research must shift from extracting data from marginalized populations to empowering them as partners—a lesson learned from the exploitation of the Havasupai Tribe’s DNA in the early 2000s [15]. The future of medicine belongs not to the best-studied populations, but to the best science—and that science must include us all.
References: 1. Solaimani, E. (2024, September 12). History Shapes Genes: The Impact of the Founder Effect on Jewish Populations. Jnetics. https://www.jnetics.org/the-founder-effect/ 2. Carress, H., Lawson, D. J., & Elhaik, E. (2021). Population genetic considerations for using biobanks as international resources in the pandemic era and beyond. BMC Genomics, 22(1), 351. https://doi.org/10.1186/s12864-021-07618-x 3. Pollak, M. R., & Friedman, D. J. (2023). APOL1 and APOL1-associated kidney disease: A common disease, an unusual disease gene—Proceedings of the Henry Shavelle Professorship. Glomerular Diseases, 3(1), 75-87. https://doi.org/10.1159/000529227 4. Limdi, N. A., Arnett, D. K., Goldstein, J. A., Beasley, T. M., McGwin, G., Adler, B. K., & Acton, R. T. (2008). Influence of CYP2C9 and VKORC1 on warfarin dose, anticoagulation attainment and maintenance among European-Americans and African-Americans. Pharmacogenomics, 9(5), 511-526. https://doi.org/10.2217/14622416.9.5.511 5. Dean, L., & Kane, M. (2012, March 8). Clopidogrel therapy and CYP2C19 genotype. In V. M. Pratt, S. A. Scott, M. Pirmohamed, et al. (Eds.), Medical Genetics Summaries. National Center for Biotechnology Information (US). https://www.ncbi.nlm.nih.gov/books/NBK84114/ 6. Dean, L., & Kane, M. (2012, September 20). Codeine therapy and CYP2D6 genotype. In V. M. Pratt, S. A. Scott, M. Pirmohamed, et al. (Eds.), Medical Genetics Summaries. National Center for Biotechnology Information (US). https://www.ncbi.nlm.nih.gov/books/NBK100662/ 7. González, M. J., Reyes, M., & Lera, L. (2021). Genetic variants in the SLC16A11 gene are associated with increased BMI and insulin levels in nondiabetic Chilean population. Archives of Endocrinology and Metabolism, 65(3), 351-358. https://doi.org/10.20945/2359-3997000000359 8. Guella, I., Asselta, R., Ardissino, D., Merlini, P. A., Peyvandi, F., Kathiresan, S., Mannucci, P. M., Tubaro, M., & Duga, S. (2010). Effects of PCSK9 genetic variants on plasma LDL cholesterol levels and risk of premature myocardial infarction in the Italian population. Journal of Lipid Research, 51(11), 3342-3349. https://doi.org/10.1194/jlr.M010009 9. Dinh, K. M., Kaspersen, K. A., Mikkelsen, S., Kjerulff, B. D., Boldsen, J. K., Petersen, M. S., Burgdorf, K. S., Sørensen, E., Aagaard, B., Forman-Ankjær, B., Bruun, M. T., Banasik, K., Hansen, T. F., Nyegaard, M., Rohde, P. D., Brunak, S., Hjalgrim, H., Ostrowski, S. R., Pedersen, O. B., Ullum, H., Erikstrup, L. T., & Erikstrup, C. (2024). Impact of CCR5Δ32 on the risk of infection, Staphylococcus aureus carriage, and plasma concentrations of chemokines in Danish blood donors. eBioMedicine, 109, 105406. https://doi.org/10.1016/j.ebiom.2024.105406 10. Inlamea, O. F., Soares, P., Ikuta, C. Y., Heinemann, M. B., Achá, S. J., Machado, A., Ferreira Neto, J. S., Correia-Neves, M., & Rito, T. (2020). Evolutionary analysis of Mycobacterium bovis genotypes across Africa suggests co-evolution with livestock and humans. PLOS Neglected Tropical Diseases, 14(3), e0008081. https://doi.org/10.1371/journal.pntd.0008081 11. Mohammed, A. O., Attalla, B., Bashir, F. M., Ahmed, F. E., El Hassan, A. M., Ibnauf, G., Jiang, W., Cavalli-Sforza, L. L., Karrar, Z. A., & Ibrahim, M. E. (2006). Relationship of the sickle cell gene to the ethnic and geographic groups populating the Sudan. Community Genetics, 9(2), 113-120. https://doi.org/10.1159/000091489 12. Damrauer, S. M., Chaudhary, K., Cho, J. H., Liang, L. W., Argulian, E., Chan, L., Dobbyn, A., Guerraty, M. A., Judy, R., Kay, J., Kember, R. L., Levin, M. G., Saha, A., Van Vleck, T., Verma, S. S., Weaver, J., Abul-Husn, N. S., Baras, A., Chirinos, J. A., Drachman, B., Kenny, E. E., Loos, R. J. F., Narula, J., Overton, J., Reid, J., Ritchie, M. D., Sirugo, G., Nadkarni, G., Rader, D. J., & Do, R. (2019). Association of the V122I hereditary transthyretin amyloidosis genetic variant with heart failure among individuals of African or Hispanic/Latino ancestry. JAMA, 322(22), 2191-2202. https://doi.org/10.1001/jama.2019.17935 13. All of Us Research Program. (2024, February 19). 275 million new genetic variants identified in NIH precision medicine data. National Institutes of Health. https://www.nih.gov/news-events/news-releases/275-million-new-genetic-variants-identified-nih-precision-medicine-data 14. Wang, L., Scherer, S. E., Bielinski, S. J., Muzny, D. M., Jones, L. A., Black, J. L., … Weinshilboum, R. M. (2022). Implementation of preemptive DNA sequence-based pharmacogenomics testing across a large academic medical center: The Mayo-Baylor RIGHT 10K Study. Genetics in Medicine, 24(5), 1062-1072. https://doi.org/10.1016/j.gim.2022.01.022 15. Kabata, F. (2024). Indigenous Peoples’ human genomic sovereignty: Lessons for Africa. Developing World Bioethics. https://doi.org/10.1111/dewb.12466
Essays Honourable Mention 5
Genetic Diversity: A Key Role in Global Health Access and the Fight Against Disease
YİĞİT JENAR YEŞİLYURT ODTÜ GELISTIRME VAKFI ÖZEL KAYSERI ANADOLU LISESI, Kocasinan, Turkey
Over the past century, numerous studies have helped humanity understand how and why genetic diversity arises. The foundations of genetics were laid by James Watson and Francis Crick, the behaviors of genetic diversity were explored by Barbara McClintock, and perhaps most relevant to this essay, the CRISPR-Cas9 technology was developed by Jennifer Doudna and Emmanuelle Charpentier. These scientific milestones have significantly contributed to our current understanding of genetics. Throughout the history of genetic research, it has been observed that certain ethnic groups and animal species possess natural resistance to specific diseases. For instance, people of Viking ancestry from Northern Europe are believed to have a natural immunity to HIV due to a mutation in the CCR5 gene (Δ32), which prevents the virus from entering cells. Such examples are not limited to humans; they are also observed in other animals. Elephants, which have more cells and therefore a theoretically higher risk of cancer, carry twenty copies of the TP53 gene, while humans have only one. This gene acts as a tumor suppressor by triggering the death of cells with high DNA damage. These cases underline the critical importance of genetic diversity in disease resistance. Genetic diversity also explains why individuals respond differently to various diseases. For example, mutations in the BRCA1 and BRCA2 genes significantly increase the risk of breast and ovarian cancer, particularly in women of Western European descent. However, these genetic variants do not occur at the same frequency in all ethnic groups, making it essential to consider diversity when designing universal genetic screening programs. Ignoring such differences may lead to overlooking vital risks in underrepresented communities. Sickle cell anemia is another example tied to genetic diversity. This disease results from a mutation in the HBB gene and is more common among individuals of African descent. The high frequency of this mutation in malaria-endemic regions illustrates an evolutionary advantage—individuals with one copy of the allele are more resistant to malaria. However, individuals with two copies face severe health consequences. This example demonstrates how genetic diversity influences not only disease susceptibility but also adaptation to environmental pressures. Tay-Sachs disease provides a further case. Caused by mutations in the HEXA gene, this fatal neurodegenerative disorder is notably prevalent among genetically isolated populations. Such examples emphasize the necessity for community-specific genetic screening programs to detect and manage hereditary conditions effectively. Moreover, the majority of today’s pharmaceutical drugs are developed based on clinical trials involving populations with relatively homogeneous genetic backgrounds, often of European ancestry. As a result, individuals with different genetic makeups may experience unexpected side effects or fail to respond to treatments. Variations in the CYP450 gene family, which influences how the liver metabolizes drugs, underscore this problem. Genetic differences in these enzymes necessitate personalized dosing and have become a cornerstone of the emerging field of personalized medicine. Despite the growing awareness of this issue, large-scale biobanks such as the UK Biobank, Estonian Biobank, and FinnGen still disproportionately represent European populations. This imbalance highlights a significant gap in the genetic data of people from Africa, Asia, and South America. Such underrepresentation risks perpetuating global health inequalities and limits our understanding of disease susceptibility in diverse communities. Thus, the establishment of more inclusive and equitable genetic databases is both a scientific and ethical imperative. In conclusion, genetic diversity plays a crucial role in shaping both health and disease outcomes. Focusing genomic research solely on specific populations can compromise scientific accuracy and social justice. The future of genetics lies in comprehensive and inclusive studies that reflect the full spectrum of human genetic variation. Only through this approach can we develop truly effective treatments and ensure personalized, equitable healthcare for all.
References: • McClintock, B. (1950). The origin and behavior of mutable loci in maize. PNAS, 36(6), 344-355. https://doi.org/10.1073/pnas.36.6.344 • Doudna, J. A., & Charpentier, E. (2014). The new frontier of genome engineering with CRISPR-Cas9. Science, 346(6213), 1258096. https://doi.org/10.1126/science.1258096 • Hütter, G., et al. (2009). Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. NEJM, 360, 692-698. https://doi.org/10.1056/NEJMoa0802905 • Abegglen, L. M., et al. (2015). Potential Mechanisms for Cancer Resistance in Elephants. JAMA, 314(17), 1850-1860. https://doi.org/10.1001/jama.2015.13134 • Piel, F. B., et al. (2010). Global distribution of the sickle cell gene. Nat Comm, 1, 104. https://doi.org/10.1038/ncomms1104 • Sirugo, G., Williams, S. M., & Tishkoff, S. A. (2019). The Missing Diversity in Human Genetic Studies. Cell, 177(1), 26-31. https://doi.org/10.1016/j.cell.2019.02.048
Essays Honourable Mention 6
KRAS: Undruggable to Treatable
Katie Longley The Wallace High School, Lisburn, UK
Understanding genetic diversity is vital to combatting disease as it is used in ‘Precision Medicine’, a medical approach in which an individual’s unique genetic makeup is taken into account when treating their illness. 5 It contrasts the ‘one size fits all’ approach within medicine by taking into account inter-patient genetic variability and tailoring their treatment accordingly. 5 Our genome is a unique set of genetic information composed of 3 billion base pairs of DNA. 1 It has been estimated that the human genome has approximately 20,000 ‘genes’, with the majority of genes acting as a set of instructions to make proteins 2. Genetic diversity is the total variety of genes within a species, and as humans, no two individuals (bar identical twins) are genetically identical 1. The total differences in the DNA of all individuals in a species form the foundations of the genetic diversity of that species. Genetic diversity stems from differences in DNA sequences between individuals. 4,KL The DNA double helix is built from four different nucleotides, each assigned a letter corresponding to the nitrogenous base it contains: adenine (A), thymine (T), guanine (G) or cytosine (C). 3 Polymorphisms (genetic differences) most commonly originate from base pair differences, referred to as ‘Single Nucleotide Polymorphisms’, which occur approximately once in every 1000 bases 1. An alteration to even a singular base in a gene could give rise to a slightly different codon, coding for a potentially different amino acid, which may alter the properties of the encoded protein. It is these differences that give rise to genetic variation. If a variation is beneficial, it will accumulate in the population; if it is disadvantageous, it will be lost. 1, KL The benefits of understanding genetic diversity can be seen through the treatments of cancer caused by KRAS, which is the most frequent oncogene, present in an estimated 25% of tumours. 8,9 Globally, cancer remains the leading cause of death, however its treatments and outcomes have been significantly reshaped and improved via the use of ‘targeted therapies’. These are a specific type of precision medicine, involving an understanding of genetic diversity, and have revolutionised the treatment of cancers caused by a KRAS mutation. 8 In its non-mutated ‘wild type’ form, KRAS, a gene on the short arm of chromosome 12, encodes for the KRAS protein, a key controller to multiple signalling pathways, regulating cell growth. 7,8,9 These proteins act as a special type of ‘on-off switch’, being activated by growth factors such as ‘receptor tyrosine kinase’. 8,9 This is reflected in its regulation of the ‘P13-AKT-mTOR’ pathway, which itself is a key regulator of cellular processes including proliferation, differentiation and apoptosis 8. However, mutations in this gene can occur, most commonly originating from single-base ‘missense mutations’; 98% of the time these base differences occur at codon 12, 13 or 61. 8 KRAS mutations will cause KRAS to become stuck in the activated position, leading to uncontrollable cell division, causing abnormal cancerous growth. 10 Oncologists are now able to determine if someone has a KRAS mutation in their tumour by carrying out a ‘genetic sequencing’ on the patient’s tumour tissue 9. Where understanding genetic diversity become important is determining which genetic mutation of KRAS is present; specifically, KRAS G12C, KRAS G12D and KRAS G12V mutant cancers will all need different treatments. 6 In recent years, drugs have been developed to directly target and block G12C mutant KRAS, with these more targeted therapies offering breakthroughs in cancer treatment. Previously, mutant KRAS was thought to be ‘undruggable’ 9. Sotorasib is the most advanced KRAS G12C inhibitor; it binds to the cysetine amino acid to essentially ‘switch off’ the mutated protein, inhibiting cell proliferation 10. However, the G12D mutation would not be effectively treated with this drug, as it doesn’t have the cysteine residue that the Sotorasib can bind to, and instead must be treated with different specific drug, MRTX1133, which targets the aspartate (D) residue 6. There are however no drugs yet developed for other G12 KRAS mutants,like G12V. To conclude, targeted therapies have shown an 80% success rate in action, proving that an understanding of the genetic diversity of tumours in a population of cancer patients is effective, and can be used to tailor treatments for individual patients based on the specific mutations in their cancer 11,7, KL. This ‘Precision Medicine’ approach, incorporating an understanding of genetic diversity, is the future of medicine, which can clearly be seen through ‘KRAS’- a once deemed ‘undruggable’ mutation now made treatable KL.
References:
(1) National Library of Medicine “Understanding Human Genetic Variation” (published 2007) ncbi.nlm.nih.gov/books/NBK20363/ (2) National Library of Medicine, Medline Plus “What is a gene?” (last updated May 2024) medlineplus.gov/genetics/understanding/basics/gene (3) Scitable, nature.com “Ribosomes, Transcription and Translation” (published 2014) nature.com/scitable/topicpage/ribosomes-transcription-and-translation-14120660 (4) Frontiers for Young Minds “What is genetic diversity?” (January 2021) kids.frontiersin.org/articles/10.3389/frym.2021.656168 (5) NHS, Transformation Directorate “Precision Medicine” (2020) transform.England.nhs.uk/ai-lab/explore-all-resources/understand-ai/precision-medicine/ (6) Personal Communications (7) National Library of Medicine, Medline Plus “KRAS Gene” (December 2017) medlineplus.gov/genetics/gene/kras/#conditions (8) nature.com “KRAS mutation: form undruggable to druggable in cancer” (November 2021) nature.com/articles/s41392-021-00780-4 (9) MD Anderson Cancer Centre “Targeting the KRAS mutation for more effective cancer treatment” (2021) mdanderson.org/cancerwise/targeting-the-KRAS-mutation-for-more-effective-cancer-treatment (10) National Library of Medicine “Classification of KRAS activating mutations and the implications for therapeutic intervention” pms.ncb.nlm.nih.gov/articles/PMC8988514/#S4 (11) SamitiveJ “Targetted Therapy; stopping cancer in its tracks” (March 2018) samitivejhospitals.com/article/detail/stopping-cancer-targeted-therapy