Exploring Medicine & Biochemistry: Research & Insights
Stay informed on the advancing science behind medical and biochemical discoveries, through well-researched articles created to inform, educate and inspire. Founded by me, Arina Kozerog, this website serves as a knowledge hub for comprehensive summaries of complex biochemical processes, insightful analysis of scientific research and latest advancements in healthcare. Whether you’re a fellow student, aspiring scientist or just curious about medicine, you’ll find engaging content that bridges biochemical concepts with real-world applications in human health and treatment. This website is a demonstration of a deep love for science and a commitment to sharing knowledge with a broader audience.
Regenerative Medicine: can we grow new organs?
The Science Behind Cancer Therapies: from chemotherapy to CAR-T and more
Fun facts about me:
Hi, I’m Arina!
I am an IB student at Cheltenham Ladies’ College with a passion for medicine, biochemistry and research.
I enjoy
Investigating different fields, from economics to neuroscience, and finding connections between them.
I am happiest
When I am active. I’ve recently picked up running and am training to run a half-marathon soon!
Explorer of knowledge: from summer programs in the US to clinical experiences in Europe, I love crossing borders in learning and life.
I’m a global
I naturally
like taking the lead when it helps the group move forward, but I think good leadership is more about listening and adapting than being in charge.
Here, I plan to share my knowledge and research with other students and everyone eager to learn more about science!

Apart from articles on various topics, you will find thorough overviews of my other projects, interviews with fascinating people in the STEM community, and an opportunity to gain study resources and 1:1 tutoring from me or other exceptional students. My articles aim to simplify complex ideas into a digestible format for someone new to the concept, it will be my writing alongside some guest contributions. Additionally, I will share my journey through competitions, research and reflections on work experience. The type of people you can expect interviews from coming soon are CLC alumni in medicine / STEM, researchers, doctors and fellow students.
Articles
Unfolding the Mystery of Proteins:
Structure, Function and Impact
The elegance of protein design
Proteins are the molecular foundations of life! They do everything from speeding up chemical reactions to giving our cells their shape and structure. But what makes them so versatile? It all comes down to their unique structure, which determines how they function. In this article, I’ll explore how proteins are built, how their structure relates to their function, and why even small changes can have dramatic effects.

The building blocks: amino acids and peptide bonds
All proteins are composed of amino acids, a monomer that links together to form polypeptide chains. There are 20 standard amino acids, each with a central carbon (the alpha carbon), an amino group (-NH2), a carboxyl group (-COOH), and a variable R group. It’s the R group that determines an amino acid’s properties — whether it’s hydrophilic, hydrophobic, acidic, or basic.
Proteins are remarkable molecules, balancing complexity with precision. Their structure, from simple amino acid sequences to sophisticated multi-subunit arrangements enables life’s most intricate processes. We, as scientists, continuing to study proteins, unlock deep insights into health, disease, and biotechnology, opening doors to new treatments and innovations.

Fun fact: we have all heard the paradox, ‘What came first, the chicken or the egg?’ Although this is still not definitively resolved and some scientists might not agree on one answer, from a molecular biology perspective, the egg came first! Here’s why:

The hard eggshell of a chicken egg is formed using a protein called ovocleidin-17 (OC-17), which is found only in hens. However, for a chicken to exist, it must hatch from an egg. This suggests that a proto-chicken (a bird very similar to a chicken but not quite the same species) laid an egg containing a genetic mutation that created the first true chicken. Since the egg existed before the chicken inside hatched, the egg came first!

So, proteins like OC-17 played a key role in the evolution of modern chickens - making this classic paradox a little less mysterious.
Amino acids are linked by peptide bonds through a condensation reaction, where a molecule of water is released. This linear sequence of amino acids forms the primary structure, which serves as the foundation for all higher levels of protein structure.
Fun fact: you’ve most likely heard of collagen from skincare advertising, and I am about to bust a myth. Collagen creams don’t work!! Collagen is a large, fibrous protein responsible for skin strength and elasticity. Many skincare products claim to deliver collagen topically, but there’s a catch - collagen molecules are simply too large to be absorbed through your skin. Instead of penetrating the deeper layers where they could be useful, collagen molecules remain on the surface, where you wash it off in the morning)
Primary structure
The sequence of amino acids in a polypeptide is determined by the genetic code. A single mutation in this sequence can be life-altering. For instance, let’s consider sickle cell disease, where a single amino acid substitution in haemoglobin causes the protein to form rigid fibres, distorting red blood cells and affecting their ability to function correctly.

Secondary structure
As soon as a polypeptide chain forms, it stars to fold into regular structures due to hydrogen bonding. The two most common types of secondary structures are:
  Alpha helices: coiled structures stabilised by hydrogen bonds between every fourth amino acid. Found in keratin (hair, nails) and membrane-spanning proteins.
  Beta pleated sheets: zigzag arrangements of polypeptides, held together by hydrogen bonds. Found in fibroin (silk) and many enzymes.
Levels of protein structures:
Structure dictates function: a few key examples
Transport and signalling proteins
Enzymes as biological catalysts
Enzymes speed up biochemical reactions by lowering the activation energy required for a given reaction. In terms of chemistry, this means more particles will have enough energy to perform successful collisions, increasing the rate of the reaction. Enzymes specificity arises from their unique active sites, which follow the induced fit model. The induced fit model is the accepted theory that enzymes undergo slight conformational changes to accommodate substrates. Namely, DNA polymerase, which accurately replicated DNA.

Structural proteins
  Collagen provides tensile strength to connective tissues.
  Keratin forms protective structures like skin and nails.
  Actin and myosin drive muscle contraction, showcasing how protein structure enables dynamic function.
  Haemoglobin binds and releases oxygen efficiently, thanks to its quaternary structure and allosteric regulation

  Membrane proteins like ion channels control cellular communication and homeostasis
  Insulin, a hormone, binds receptors to regulate glucose uptake
Figure 1: general amino acid structure
Though it is heartbreaking to attribute such life-altering conditions to tiny mutations in one of the simplest structures in our bodies, understanding these failures has led to breakthroughs in medicine, including drug design targeting protein structure.
The precise folding of proteins is crucial as errors can lead to devastating consequences:
  Prion diseases like Creutzfeldt-Jakob Disease occur when incorrectly folded proteins induce normal proteins to misfold, leading to brain damage, and causing dementia, involuntary muscle movements, loss of intellect and coordination, changes in personality and slurred speech. Lots of people with Creutzfeldt-Jakob Disease die within a year of symptom onset.

  Cystic fibrosis results from a mutation in the CFTR protein, which is responsible for maintaining proper hydration and mucus function in various organs, especially the lungs. This mutation disrupts ion transport in cells, resulting in thick, sticky mucus buildup in the lungs and other organs. People with cystic fibrosis often experience recurring chest infections, difficulty gaining weight, frequent coughing, and wheezing.
When protein structure fails
Tertiary structure
Quaternary structure
The tertiary structure results from further folding, driven by interactions between the R groups:

  Hydrophobic interactions push non-polar side chains to the protein’s interior.
  Ionic bonds form between charged R groups.
  Disulphide bridges are covalent bonds between cysteine amino acids, they create strong structural reinforcements.
  Hydrogen bonds contribute to stability and specificity.
The 3D shape is vital. Enzymes, for example, have active sites precisely shaped to bind substrates. Incorrect folding at this stage can lead to diseases like Alzheimer’s, where incorrectly folded proteins aggregate into plaques, interfering with brain cell function, leading to memory loss, impaired thinking, and eventually an inability to perform daily tasks.
Some proteins are multiple tertiary structures joined together, they function as multi-subunit complexes. Such as haemoglobin, which consists of four polypeptides working together to transport oxygen. The quaternary structure allows for cooperative interactions, enhancing function.
Conjugated proteins also have one or more non-protein parts to the molecule, like haemoglobin’s ‘haem’ group.
(See Figure 4)
Regenerative medicine: can we grow new organs?
Imagine a world where organ failure is no longer a life-threatening condition, and patients do not have to wait years for a transplant. Regenerative medicine, a revolutionary field combining biology, technology, and medicine, is making this vision a reality. Scientists are developing ways to repair, regenerate, and even grow new organs using stem cells, tissue engineering, and 3D bioprinting. But how does this work, and what challenges remain before lab-grown organs become a routine part of medicine?

How does regenerative medicine work?

Regenerative medicine is based on the body's ability to heal itself. By using living cells, scientists can regenerate tissues and organs, potentially eliminating the need for traditional organ transplants. The key components of regenerative medicine include:
Stem cells are the foundation of regenerative medicine because they the ability to develop into different types of cells in the body. There are three main types of stem cells used in research:

Embryonic Stem Cells (ESCs): These have the highest potential for regeneration, as they can develop into almost any type of cell, but are controversial due to ethical concerns.

Adult Stem Cells: Found in tissues like bone marrow and fat, these are limited in what they can become but are used in treatments like bone marrow transplants, or other very specific stem cell therapies.

Induced Pluripotent Stem Cells (iPSCs): Adult cells derived from skin or blood, that have been reprogrammed to behave like embryonic stem cells, eliminating ethical concerns while still offering vast potential. They sound like a miracle solution however; these aren’t widely used due to disadvantages like tumorigenesis and genetic instability. The reprogramming process used to create iPSCs can introduce genetic changes that increase the risk of tumor formation. As well as the likelihood that iPSCs may undergo mutations over time, potentially affecting their function and safety.

Stem cells are being used to repair damaged heart tissue after heart attacks, regenerate nerve cells for spinal cord injuries, and even grow patches of liver and kidney tissue.
Tissue engineering involves growing biological tissues outside the body and then implanting them in patients. This process typically includes:

  • Scaffolds: A 3D structure made of biodegradable materials or decellularized organs, essentially stuff that can break down in the body. They provide a framework for new cells to grow, something to hold onto and ground around.
  • Cells: Stem cells or specialized cells from the patient are seeded onto the scaffold.
  • Growth Factors: Chemical signals that encourage the cells to grow and develop into functional tissue. Growth factors are crucial for directing the development of the implanted cells on the scaffolds into functional, tissue-specific structures.

Scientists have successfully engineered skin, bladders, and cartilage using this approach, and research is advancing toward more complex organs like the liver and pancreas.

Fun Fact: In 2019, scientists at Tel Aviv University 3D-printed a miniature human heart using a patient’s own cells. It was complete with blood vessels, chambers, and ventricles. Though it was only the size of a cherry, it marked the first time an entire heart was printed with fully cellular and vascular components, proving that "printing" life isn’t just science fiction anymore!
Stem Cell Therapy
Tissue Engineering
Recent breakthroughs in organ regeneration
While we are not yet at the stage of printing fully functional organs, there have been several remarkable advancements:

1.Lab-grown bladders and artificial skin

Scientists have successfully engineered bladders in the lab and transplanted them into patients. These bladders were created using a patient's own cells, reducing the risk of immune rejection. Similarly, artificial skin has been developed for burn victims, helping speed up wound healing.

2.Miniature liver and kidney tissues

Researchers have created small liver and kidney tissues capable of performing some of their functions. While they are not yet complete organs, they can be used for drug testing and may one day support failing organs until a full transplant is available, saving lives of many, as about 46% of people on the transplant list die before receiving their organ, while a new person is added every 9 minutes. Even though this is a temporary solution, it can help bring down this figure significantly.

3.Heart tissue regeneration

Scientists have used stem cells to grow patches of heart tissue that can be implanted to help repair damage after a heart attack. This technology could one day eliminate the need for heart transplants. The first successful human heart transplant was performed in 1967 by Dr. Christiaan Barnard in South Africa. Today, heart transplants have a one-year survival rate of over 85%, but there still aren’t nearly enough donor hearts to meet the need, making lab-grown heart tissue a promising alternative for the future.

4.3D-printed blood vessels and corneas

Researchers have successfully 3D-printed blood vessels, an essential step toward printing entire organs. In addition, 3D-printed corneas have been developed, potentially solving the global shortage of cornea donors. It’s the kind of technology we expect to see in Sci-Fi films like the Blade Runner or The Island, but it’s happening in real life, and much sooner than we all thought.
3D bioprinting is an innovative technology that prints layers of living cells to create tissues and organ structures. It uses a printer similar to a normal 3D printer, but designed and adapted for biological material. The specialized printer uses bio-inks containing living cells and other ingredients that help support growth. The printer lays down the bio-ink layer by layer in a precise pattern, shaping it into things like blood vessels, skin, and even parts of organs. One of the best parts of 3D bioprinting is that it can be customized to match a specific patient. This means the printed tissue is more likely to be accepted by the body, lowering the chance of rejection and making treatments more personal and effective. While fully printed, transplantable organs are still being developed, this technology is already making a big impact in areas like drug testing, skin grafts, and tissue repair.
Figure 1: first 3D printed heart
Despite the exciting progress, regenerative medicine faces several significant hurdles before lab-grown organs become widely available.

Complexity of organ structure:

Organs like the liver and kidneys have incredibly intricate structures, with specialized cells performing different functions. Recreating this level of detail in the lab is extremely difficult and remains one of the biggest technical challenges in regenerative medicine.

Immune Rejection:

Even when a patient’s own cells are used, the immune system can sometimes react unpredictably. Rejection is still a risk, so we try to use gene editing tools such as CRISPR to make the cells less likely to trigger an immune response. CRISPR allows scientists to precisely cut and modify DNA. It works like molecular scissors, making it possible to change or fix genes with high accuracy.

Scientists predict that within the next few decades, regenerative medicine could completely transform how we treat organ failure. One exciting possibility is the creation of full-scale 3D-printed organs, such as functional hearts, kidneys, and lungs ready for transplantation. Alongside this, gene-edited regenerative therapies using tools like CRISPR may allow scientists to correct genetic defects before growing the organ, making treatments even more personalized and effective. Artificial intelligence could also play a key role by streamlining and improving the design and growth process of lab-made tissues and organs, bringing these futuristic treatments closer to reality. AI has the ability to optimise our current approach to regenerative medicine and personalise it further to each patient.

If these advancements continue, we could one day live in a world where no one has to wait, sometimes for years, for a life-saving organ transplant. That would be a game-changer for millions of people. Right now, far too many patients die simply because a compatible donor isn’t found in time. But with lab-grown organs, there could be a reliable, endless supply, giving people the second chance they desperately need. It would also take pressure off the donation system and make transplants safer and more successful. Since these organs could be made from a patient’s own cells, the risk of rejection would be much lower, and many people might not need to take harsh immunosuppressive drugs for the rest of their lives.
Regenerative medicine is one of the most promising fields in modern science, offering hope for patients suffering from organ failure. From stem cell therapy to 3D bioprinting, researchers are developing groundbreaking technologies that could revolutionize healthcare. While challenges remain, ongoing advancements suggest that a future, where we can grow new organs from a patient’s own cells is not just possible in science fiction, it’s on the horizon for us.
Ethical concerns

  • Embryonic stem cells: The use of ESCs remains controversial because they come from human embryos, and from a philosophical standpoint, do you feel the question of whether an embryo is alive or not will ever be answered? Does it have a right to informed consent and autonomy like adult human beings? Does the parent have a right to dictate its fate?
  • Designer organs: With the possibility of genetic engineering and DNA modification a new question of ethics rises: If we can grow new organs, should we enhance them beyond natural limits?
  • Accessibility and cost: Such medical breakthroughs can take decades to become accessible to all and for the foreseeable future, who will have access to these treatments, and will they be affordable, provided by public healthcare institutions?
The blood supply struggle (vascularization):

One of the biggest barriers to growing functional organs is creating an internal network of blood vessels to deliver oxygen and nutrients. Without this, large tissues cannot survive after implantation. Scientists are making progress, but fully functional vascular systems in lab-grown organs are still hard to achieve.

Challenges and ethical considerations
The future of regenerative medicine
3D-Bioprinting
The Rise of Telemedicine: benefits and challenges of virtual healthcare
During the COVID-19 pandemic, when hospitals were overwhelmed and lockdowns kept people at home, healthcare had to find a new path forward. That path was telemedicine, the remote delivery of healthcare using digital technology. What began as a temporary response has since become a permanent fixture in many healthcare systems.

Telemedicine promises to make healthcare more accessible, efficient, and patient-centred. But while its benefits are clear, its rise also brings new challenges around equity, privacy, regulation, and clinical care.
Telemedicine’s roots can be traced back to the 1950s, when hospitals began experimenting with tele-radiology and video consultations. However, adoption remained limited due to technological and regulatory barriers.
1960-70s: early experiments

  • 1960s (USA): NASA begins experimenting with remote monitoring of astronauts’ health in space, laying the foundational technology for telemedicine

  • 1967: Massachusetts General Hospital establishes a telemedicine link with Logan International Airport to provide medical consultations to travellers

  • 1970s: Telehealth pilot programs launch in rural parts of Canada and Alaska to reach remote communities
1980s: slow development

  • 1980s: The introduction of personal computers and early video conferencing allowed for experimentation with teleconsultations, but high costs and a lack of internet limited widespread adoption

  • Some hospitals began transmitting radiologic images electronically, an early form of tele-radiology.
1990s: Internet brings expansion

  • 1990s: The growth of the internet and email made remote communication between doctors and patients more feasible.

  • 1997: American Telemedicine Association (ATA) was founded to promote the development and use of telehealth services.

  • Late 1990s: Virtual consultations gained traction in dermatology and mental health, especially in rural clinics.


2010–2019: mainstream growth

  • 2010s: Cloud computing, wearable devices, and secure patient portals expand the potential for virtual care

  • 2015: Medicare (US) expanded coverage for some telehealth services

  • 2016–2019: Global telemedicine market grows rapidly, with rising investment in AI diagnostics, remote monitoring, and app-based healthcare platforms
2000s: technology matures

  • Early 2000s: Broadband expansion, better webcams, and digital records make virtual care more reliable

  • 2005: The US Department of Veterans Affairs began large-scale telehealth programs, especially for chronic condition management

  • 2008: Smartphones and health apps emerged, enabling basic remote monitoring and communication.
2022–present: integration and innovation


  • 2022–2024: Hybrid healthcare models emerge: patients choose between in-person and virtual visits

  • AI and wearable tech begin playing major roles in diagnostics, triage, and monitoring

  • 2025: Global policy reform begins to catch up with telehealth’s growth, though challenges remain in regulation and equity
Fun Fact: In the near future, VR could allow doctors to conduct immersive remote surgeries or simulate patient examinations in 3D. Imagine a surgeon in London can ‘step into’ a virtual operating room with a patient across the globe, all without leaving their office. Soon, you will be able to chat with your physician, point to what hurts on a 3D version of yourself, or even do rehab exercises, all in a VR environment/gym, without changing out of your pj’s.
A brief history + timeline:
2020: climax

  • March 2020: COVID-19 pandemic prompts governments to lift many restrictions on virtual care

  • 2020–2021: Services expand across primary care, mental health, and chronic disease management

  • Insurance providers and health systems rapidly scaled up virtual platforms and integrated telemedicine into routine care

The COVID-19 pandemic acted as a catalyst, triggering an explosion in telemedicine adoption. In the US, telehealth usage jumped more than 1500% in early 2020. Countries across Europe and Asia saw similar shifts, with healthcare providers rapidly moving consultations online.
Today, telemedicine encompasses a wide range of services that extend far beyond simple video calls. It includes general medical consultations, remote management of chronic diseases, virtual mental health services, AI-powered symptom checkers, and wearable health tracking with real-time remote monitoring. These services are made possible by a growing ecosystem of technologies designed to streamline and enhance the delivery of virtual care. Secure video conferencing platforms, such as Zoom for Healthcare and Doxy.me, form the backbone of many remote consultations. Mobile apps allow patients to book appointments, receive prescriptions, and access test results with ease. Wearable devices track vital signs in real time, feeding data directly to healthcare providers, while Electronic Health Records (EHRs) ensure seamless access to patient history. Additionally, artificial intelligence is increasingly used for triaging symptoms and supporting diagnostic decisions, helping to improve both the speed and accuracy of virtual care.
Increased Accessibility
Telemedicine removes geographic and logistical barriers. Rural patients, elderly individuals, and those with disabilities can consult doctors without needing to travel long distances. Virtual care also benefits people in underserved areas with physician shortages.

Cost-Effectiveness
Telehealth reduces expenses for both patients and providers. Patients save on transportation and lost wages, while clinics reduce overhead costs. Fewer unnecessary ER visits and hospital readmissions also ease pressure on healthcare systems, which has been seen to have been especially effective in the UK, where the public National Health Service (NHS) reported a 35% reduction in hospital admissions for certain conditions managed through virtual consultations.

Time Efficiency
Appointments can be scheduled quickly and flexibly, with minimal waiting room time. 24/7 services allow patients to get care outside of typical office hours, increasing convenience and responsiveness.

Continuity of Care
Telemedicine enables consistent follow-ups for chronic conditions like diabetes or hypertension. Patients can communicate easily with their healthcare teams and access test results, prescriptions, and monitoring from one platform.

Mental Health Support
Virtual therapy and counselling have become mainstream, especially among young adults. Remote access helps reduce stigma, increases privacy, and shortens wait times for mental health services.
The Digital Divide
Access to high-speed internet, smartphones, and digital literacy is uneven. Elderly patients and low-income populations are most at risk of being left behind, deepening health disparities. This is a huge limitation, as these services are designed to benefit these same groups of people, and there is a high proportion of them that will have difficulty accessing telemedicine.

Privacy and Security
Digital health data is vulnerable to cyberattacks and breaches. Ensuring secure communication and storage in compliance with regulations like HIPAA (US) and GDPR (EU) is essential, yet often difficult. Scepticism may also undermine patients' confidence that their conversations with medical professionals remain confidential, potentially affecting trust in the preservation of doctor-patient confidentiality laws.

Clinical Limitations
Virtual care cannot fully replicate physical examinations. Certain diagnoses, tests, and treatments still require in-person visits, which limits telemedicine’s applicability for complex or emergency cases.

Regulatory and Licensing Issues
Healthcare is regulated locally, but telemedicine often crosses borders. Doctors may face legal restrictions treating patients in other states or countries. Licensing and telehealth laws vary widely, creating a patchwork of rules.

Reimbursement and Insurance
Insurance coverage for telehealth services is inconsistent. Some providers don’t reimburse at parity with in-person care, creating financial disincentives for practitioners and confusion for patients.
Looking ahead, I believe telemedicine is in a position to become an integral part of modern healthcare rather than a temporary solution born of crisis. The future of virtual care will likely be shaped by the continued integration of artificial intelligence, wearable health technology, and advanced remote monitoring systems. AI has the potential to revolutionize diagnostics, triage, and personalized treatment planning, while wearables, such as smartwatches and biosensors, can provide real-time data on vital signs, enabling clinicians to track patients’ health remotely with greater accuracy. Alongside these advancements, hybrid models of care are expected to become the norm, blending the convenience of virtual consultations with the necessity of in-person diagnostics, physical exams, and procedures. To support this evolution, significant policy reforms will be needed. These include establishing universal standards for data privacy and security, simplifying cross-border licensing for clinicians, and ensuring fair and consistent reimbursement structures across public and private systems. Importantly, efforts must also be made to close the digital divide by investing in broadband infrastructure and digital literacy programs, especially for underserved communities. Telemedicine also offers an underappreciated environmental advantage: by reducing patient and clinician travel, it lowers carbon emissions and contributes to a more sustainable healthcare model. If embraced responsibly and inclusively, the future of telemedicine holds enormous promise for delivering smarter, safer, and more equitable care around the world.
Telemedicine has reshaped the way we think about healthcare, turning homes into clinics and devices into diagnostic tools. Its benefits are transformative: greater accessibility, lower costs, and improved patient engagement. But its challenges, especially digital inequality, privacy risks, and regulatory gaps, must be addressed.

As technology and policy evolve, telemedicine can become more than a pandemic-era stopgap. It can be a permanent, powerful complement to in-person care. Hopefully, the rise of telemedicine will contribute towards building a smarter, more inclusive future for global health.
Figure 1: Key benefits of telemedicine, as identified by Pexip.com
Figure 2: Use of virtual reality in healthcare
Benefits of telemedicine
Challenges and limitations of telemedicine
The Science Behind Cancer Therapies
Cancer, a complex group of diseases, is caused by uncontrolled cell division. It is often characterized by the potential to invade or metastasize to other tissues, and remains one of the leading causes of death worldwide. The heterogeneity of cancer, encompassing hundreds of distinct subtypes with varied molecular profiles, presents immense challenges for effective treatment. However, over the past century, we have witnessed an evolution of cancer therapy, from crude cytotoxins to precision-guided biological interventions, this has transformed patient outcomes in many contexts. In this article, I’ll delve into the science of major current cancer treatments, including chemotherapy, radiation, immunotherapy, and CAR-T cell therapy. I’ll also examine how genetic and lifestyle factors contribute to cancer risk, and highlight promising avenues in ongoing research.
Chemotherapy refers to the systemic administration of cytotoxic agents that target rapidly dividing cells. Most chemotherapeutic drugs, such as alkylating agents (e.g., cyclophosphamide), antimetabolites (e.g., 5-fluorouracil), and anthracyclines (e.g., doxorubicin), act by interfering with DNA replication or mitotic processes, where the uncontrolled division stems from. These agents are non-specific, affecting both cancerous and normal proliferative cells, notably those in the gastrointestinal tract, bone marrow, and hair follicles. This lack of selectivity accounts for common side effects such as immunosuppression, mucositis, alopecia, and nausea. Resistance to chemotherapy, through mechanisms such as increased drug efflux (e.g., P-glycoprotein expression) or DNA repair upregulation, remains a central barrier to efficacy and often drives treatment failure in relapsed cancers.
Radiation therapy (RT) uses ionizing radiation, X-rays or gamma rays, to induce double-stranded DNA to break within tumour cells, leading to apoptosis or mitotic catastrophe, two forms of cell death. There are two primary delivery methods: external beam radiation therapy (EBRT) and brachytherapy, where radioactive sources are placed close to or within the tumour. RT is especially effective for localized tumours and is often employed in adjuvant or palliative settings, where RT is employed after a primary treatment to avoid reoccurrence of cancer or when the treatment is used in terminal patients to avoid spread and improve the quality of life left. The biological effectiveness of RT is modulated by fractionation, oxygenation status (hypoxic tumours are more resistant), and the DNA repair capacity of the targeted cells. Advances such as intensity-modulated radiation therapy (IMRT) and proton therapy have improved the spatial precision of dose delivery, minimizing collateral damage to surrounding healthy tissue.
Immunotherapy represents a paradigm shift in cancer treatment by using the patient’s immune system to detect and destroy malignant cancerous cells. One of the foundational strategies involves immune checkpoint inhibitors (ICIs), which target negative regulators of T-cell activation, such as PD-1, PD-L1, and CTLA-4. These molecules are often exploited by tumour cells to evade immune detection. Blocking them can unleash a potent antitumour immune response, particularly in immunogenic cancers like melanoma and non-small cell lung cancer. However, only a subset of patients exhibit durable responses, and immune-related adverse events (irAEs) ranging from dermatitis to autoimmune colitis reflect the fine balance between tumour suppression and immune tolerance.
Chimeric Antigen Receptor T-cell (CAR-T) therapy is one of the most promising forms of adoptive cell transfer in oncology. In this technique, T-cells are extracted from a patient and genetically modified ex vivo, meaning outside of the patient, to express synthetic receptors that recognize specific tumour-associated antigens, such as CD19 in B-cell cancer growths. Upon injection, these engineered T-cells find and destroy cancer cells using the target antigen. Clinical trials and approved therapies, such as tisagenlecleucel and axicabtagene ciloleucel, have demonstrated profound responses in refractory acute lymphoblastic leukemia and diffuse large B-cell lymphoma. However, challenges such as cytokine release syndrome (CRS), neurotoxicity, and limited efficacy in solid tumours persist. Ongoing research in this field, focuses on improving CAR constructs, optimizing antigen selection, and modulating the tumour microenvironment to enhance T-cell infiltration and persistence.
Patient Spotlight: Emily Whitehead, the first paediatric acute lymphocytic leukemia patient successfully treated with CAR-T therapy, has been in remission since 2012. She was only 6 when she started battling cancer, and now lives a cancer-free life and has become an advocate for paediatric cancer research and treatment. She has since twice participated in the Olympics, in 2020 and 2024, for artistic gymnastics. Her case catalyzed widespread research into T-cell engineering and led to the first FDA-approved CAR-T therapy.
The science behind cancer treatments: from chemotherapy to CAR-T and more
Chemotherapy
Radiation Therapy
Immunotherapy
CAR-T Cell Therapy
Genetic Influence on Cancer Risk
Lifestyle and Environmental Factors
Personalized Medicine
Cancer vaccines
Nanomedicine
Artificial Intelligence and clinical data
Cancer arises from accumulated genetic alterations that disrupt normal cell cycle regulation, apoptosis, and genomic stability. Inherited mutations in tumour suppressor genes, such as BRCA1/2 (breast and ovarian cancer) and TP53 (Li-Fraumeni syndrome), significantly elevate lifetime cancer risk. These germline mutations, mutations in reproductive cells, are complemented by somatic mutations, mutations in non-reproductive cells, acquired over time due to environmental insults or replication errors. Advances in next-generation sequencing have uncovered common cancer-causing mutations, like those in the KRAS, EGFR, and BRAF genes, as well as major DNA rearrangements, such as the BCR-ABL fusion. These genetic changes not only fuel tumour growth but also offer specific targets for treatment. In addition, changes in gene regulation, like DNA methylation or modifications to histone proteins, proteins that provide structural support to chromosomes by packaging and organizing DNA, can disrupt normal cell function and lead to cancer, even without altering the DNA code itself.
While genetics plays a critical role, lifestyle and environmental exposures substantially influence cancer risk. Tobacco use remains the most preventable cause of cancer worldwide, strongly associated with lung, head and neck, and bladder cancers. Diets high in red and processed meats, alcohol consumption, physical inactivity, and obesity have all been linked to increased risk through mechanisms involving chronic inflammation, hormonal imbalances, and oxidative stress. Infectious agents such as human papillomavirus (HPV), hepatitis B and C viruses, and Helicobacter pylori are connected to cervical, liver, and gastric cancers, respectively. Public health interventions, like vaccination programs (e.g., HPV vaccine), smoking reduction campaigns, and cancer screening, have significantly reduced the burden of preventable cancers. Smoking rates in the UK have significantly declined since the link between smoking and lung cancer was established in the 1950s. In the early 1950s, approximately 80% of men and 40% of women in the UK were smokers. By 1998, these figures had dropped to 26% for men and 23% for women. This decline is attributed to increased awareness of the health risks associated with smoking, public health campaigns, and tobacco control policies. Although even today these figures aren’t at 0, as a society we’ve made huge progress. Due to the addictive nature of nicotine, we cannot expect these statistics to drop any lower soon.
Personalized or precision oncology aims to tailor therapy based on the molecular characteristics of an individual’s tumour. Gene profiling can identify mutations (e.g., EGFR mutations in lung adenocarcinoma or HER2 amplification in breast cancer) that guide the use of targeted therapies such as tyrosine kinase inhibitors and monoclonal antibodies. Liquid biopsies, which detect circulating tumour DNA (ctDNA) in the blood, are emerging tools for real-time monitoring of treatment response and minimal residual disease. Personalized approaches enhance efficacy and reduce unnecessary toxicity, although challenges remain in addressing tumour heterogeneity and resistance mechanisms. Dr. James P. Allison, Nobel Laureate in Physiology and Medicine 2018, discussed the future of combining immunotherapy with precision cancer therapeutics to design individualized therapies in an interview with the American Association for Cancer Research (AACR). He emphasizes the importance of combining different therapeutic approaches to tailor treatment to individual patients.
Brief cancer treatment timeline:

  • 1890s: Surgical excision of tumors
  • 1940s: Introduction of nitrogen mustards (early chemotherapy)
  • 1950s: Development of radiation therapy machines
  • 1970s: Discovery of oncogenes
  • 2000s: Approval of monoclonal antibodies (e.g., trastuzumab)
  • 2017: First FDA approval of CAR-T therapy
  • 2020s: Rise of AI, liquid biopsies, and personalized oncology
Cancer vaccines can be categorized as prophylactic (e.g., HPV and HBV vaccines) or therapeutic. Therapeutic vaccines aim to stimulate an immune response against established tumours by introducing tumour-associated antigens or neoantigens. Experimental vaccines, such as dendritic cell vaccines like the Sipuleucel-T for prostate cancer, are being tested on various cancer growths. Success is contingent on how well the vaccine targets a specific cancer antigen, how strongly it activates the immune system, and whether it can overcome the strategies tumours use to hide from our immune system, as to not trigger a response.
Nanotechnology in oncology uses nanoparticles to deliver chemotherapeutic drugs directly to tumour cells, improving drug solubility, stability, and bioavailability. These carriers, such as liposomes, dendrimers, and polymeric nanoparticles, can be modified to carry specific binding molecules that target cancer-specific receptors, enhancing selectivity. Approved examples include liposomal doxorubicin (Doxil) and albumin-bound paclitaxel (Abraxane). Ongoing research aims to refine nanocarriers that respond to tumour microenvironmental cues, like pH and enzymes, for controlled drug release.
AI is revolutionizing cancer diagnostics and treatment planning. Machine learning algorithms can detect subtle patterns in radiologic images and histopathology slides, improving early detection and subtype classification. Integrating multi-omics data, where different types of data, like genomics, transcriptomics, proteomics, is combined during analysis, through AI, enables the identification of biomarkers and therapy targets. Predictive modelling assists oncologists in estimating treatment responses and prognoses. Ethical considerations, such as data privacy and algorithm transparency, remain critical as AI becomes more embedded in clinical oncology.
The science of cancer treatment has progressed from nonspecific cytotoxic strategies to highly personalized interventions rooted in molecular and immunologic research. Chemotherapy and radiation remain widely used, yet are now often supplemented or replaced by immunotherapies, targeted drugs, and cellular therapies like CAR-T. Meanwhile, understanding the interplay of genetic predisposition and lifestyle factors enables more effective prevention strategies. As research continues to advance, through nanotechnology, AI, and personalized medicine, the promise of more effective, less toxic, and ultimately curative treatments becomes increasingly tangible. Continued investment in interdisciplinary research and equitable access will be key to translating these scientific breakthroughs into global cancer control. Dr. Siddhartha Mukherjee, oncologist and author of The Emperor of All Maladies, highlights “Cancer begins and ends with people. In the midst of scientific abstraction, it is sometimes possible to forget this one basic fact.” This is an important reflection and reminder of the human aspect behind the scientific endeavours in cancer treatment. In research, we must keep people, their quality over quantity of life, on the forefront of our minds.
The Gut Matrix: microbes, immunity and mood
The human gut harbours a vast and dynamic ecosystem consisting of trillions of microorganisms, collectively called the gut microbiome. Unlike you might expect, these microbial populations aren’t just there; they actively influence essential physiological systems, including digestion, immune function, and even cognitive health. The relationship between the host and its microbiota is symbiotic, meaning mutually beneficial. In this case, the host provides a nutrient-rich environment, while the microbes, in return, assist in metabolic processes, modulate immune responses, and impact neurochemical signalling. In this article, I explore how these microscopic allies shape human health, how diet and lifestyle influence microbial composition, and how advances in microbiome-based medicine are reshaping therapeutic strategies.
The gut microbiome primarily comprises bacteria, but it also contains archaea, viruses (particularly bacteriophages), fungi, and protists. Within the bacterial domain, the most abundant phyla are Firmicutes and Bacteroidetes, with Actinobacteria and Proteobacteria present in smaller proportions. Everyone’s microbial fingerprint is unique, influenced by genetics, geography, early life exposures, and long-term diet. While diversity is often associated with resilience and health, emerging research suggests that the specific balance of one’s microbiome may be just as critical as overall richness.
The microbiome’s colonization begins at birth, with natural birth exposing infants to maternal vaginal and faecal microbiota, while Caesarean section often results in skin-associated microbial transmission. Breastfeeding further enriches microbial diversity via oligosaccharides that serve as prebiotic substrates for Bifidobacteria. This early microbial programming shapes immune system development and can influence disease susceptibility well into adulthood. Disruptions during these critical windows early in human life can be caused by antibiotics or poor diet and may predispose individuals to immune-related and metabolic disorders.
The human genome codes for only a limited range of enzymes to digest complex carbohydrates. The gut’s microbiota compensates for this deficit, fermenting complex polysaccharides, such as cellulose, inulin, and resistant starch, into short-chain fatty acids (SCFAs) like acetate, propionate, and butyrate. Let’s take butyrate, it serves as the primary energy source for colonocytes and exerts anti-inflammatory effects by inhibiting histone deacetylases and promoting regulatory T cell differentiation. Microbes also synthesize essential micronutrients, including vitamin K and various B vitamins, notably B12 and folate, which are otherwise poorly available.
The microbiome modulates bile acid metabolism by deconjugating primary bile acids into secondary forms, which then influences lipid digestion and cholesterol homeostasis. In the small intestine, microbial signals regulate intestinal epithelial tight junctions and mucin production, impacting gut permeability. Furthermore, bacterial metabolites such as indole derivatives from tryptophan metabolism act as ligands for aryl hydrocarbon receptors, linking microbial activity to host endocrine and detoxification pathways. Without all the science talk, gut bacteria help your body break down fats, control how much cholesterol you absorb, and maintain the lining of your gut so harmful substances don’t leak into the bloodstream. They also send signals that affect hormones and how your body handles toxins.
Around 70% of the immune system resides in the gut, particularly within gut-associated lymphoid tissue (GALT). This tissue interacts closely with the microbiome, training immune cells to tolerate commensals, the microbes that reside in our body without harming our health, while remaining responsive to pathogens, the bad microorganisms. Dendritic cells detect microbes and relay this information to naive T cells, instructing them to become either calming regulatory cells or active immune responders. When the microbial environment is balanced, it tends to favour the development of anti-inflammatory Tregs, which play a key role in preventing autoimmune diseases and allergic reactions.
Dysbiosis is the imbalance or loss of microbial diversity and plays a part in many diseases. Inflammatory bowel disease (IBD) is characterized by reduced abundance of Faecalibacterium prausnitzii, a butyrate-producing bacterium with immunoregulatory functions. Similarly, low diversity in the microbiome has been associated with increased asthma and allergy risk in children. Dysbiosis also affects systemic immunity. For example, microbiota can influence the efficacy of immunotherapy in cancer treatment, and altered gut communities are linked to increased susceptibility to infections due to impaired mucosal defences. Check out the article on the science behind cancer therapies if you want to know more about immunotherapy, and what affects its efficacy.
Figure 1: This diagram, taken from Research Gate shows the bidirectional communication between the gut, immune system, and brain, highlighting the vagus nerve, SCFAs, cytokines, and tryptophan metabolites as signalling pathways.
How do microbial communities influence digestion, immunity, and mental health
Microbial composition
Colonization and development
Enzymes
Nutrient absorption and metabolism
Gut-Assosciated Lymphoid Tissues (GALT)
Microbial dysbiosis and disease
Neurological signalling pathways
Clinical correlations
Dietary patterns
Lifestyle and environmental influence
Probiotics, prebiotics, and synbiotics
Faecal Microbiota Transplantation (FMT)
Personalized microbiome therapeutics
The gut-brain axis represents a complex communication network involving the enteric nervous system, the vagus nerve, immune mediators, and microbial metabolites. SCFAs can cross the blood-brain barrier and influence gene expression in microglia, the brain’s resident immune cells. Tryptophan, an essential amino acid metabolized by gut microbes, serves as a precursor to serotonin, 90% of which is produced in the gut. Microbes such as Lactobacillus rhamnosus can alter GABA receptor expression in the brain, modifying stress and anxiety responses. In simpler terms, the bacteria in your gut don’t just help with digestion; they send signals to your brain that can affect your mood, stress levels, and even how your brain develops. This means taking care of your gut through diet and lifestyle may help support mental wellbeing too.
Alterations in microbiome composition have been associated with neuropsychiatric conditions, including depression, anxiety, and autism spectrum disorder (ASD). Faecal microbiota transplantation (FMT) from healthy donors to individuals with major depressive disorder has shown modest improvements in mood in small clinical trials. Similarly, probiotic supplementation, often with Lactobacillus or Bifidobacterium strains, has demonstrated anxiolytic effects, reducing anxiety and some patients reported an overall effect similar to antidepressants. However, findings remain inconsistent due to the diversity in study design and strain specificity. Generally, the origins, causes, and treatments of disorders related to mental health are very unknown, and often specific to the individual patient.
Diet is a principal driver of microbiome composition. Diets high in plant-based fibers, such as the Mediterranean diet, support SCFA-producing microbes and foster microbial diversity. In contrast, Western diets high in saturated fats, simple sugars, and low in fiber are associated with pro-inflammatory microbial profiles and increased gut permeability. It is worth mentioning that transitions between dietary patterns can alter microbiota composition within 24–72 hours. This underscores its dynamic nature, and busts the idea that ‘it’s too late to change your diet’. Even though a long-term balanced diet will have the best outcome, you may notice improvements in your wellbeing within a couple of days after implementing positive dietary changes.
Beyond diet, several lifestyle factors shape microbial health. Antibiotics, while sometimes necessary and save many lives, can cause long-term shifts in microbial communities, more so if taken during early development/childhood. Chronic psychological stress alters gut motility, immune responses, and microbial diversity. Sleep deprivation and circadian rhythm disruptions also dysregulate microbial composition. These effects are seen most commonly in people who work in crazy shifts, and in commuters, who frequently experience jetlag caused by their travel. On the bright side, physical activity has been positively correlated with increased microbial diversity and elevated SCFA production. Key takeaways from this are that, where possible, you should prioritize sleep and active rest, even simply going for a walk can positively influence your gut.
Probiotics are live microorganisms that, when consumed in the right amounts, can support your health, especially gut health. You’ve probably seen the surge in probiotic supplements and drinks lining supermarket shelves, each claiming to boost digestion or immunity. Popular strains like Lactobacillus acidophilus and Bifidobacterium longum are commonly included, and they do have scientific backing in certain contexts. Prebiotics, on the other hand, are types of fiber, like inulin and FOS, that act as food for good bacteria already living in your gut. Products combining both are called synbiotics.

But despite the hype, not all probiotics live up to their promises. Many over-the-counter options don’t survive the journey through your stomach acid or fail to stick around long enough to make a real difference. Some are based on strains with little clinical evidence or include too low a dose to be effective. Take lion’s mane mushrooms, which are marketed as containing probiotics. The newfound popularity has no basis in science, as lion’s mane isn’t a probiotic in itself, and the small traces of probiotics within it have a negligible effect on our gut. That said, high-quality, strain-specific probiotics, ideally recommended by a healthcare provider, can be beneficial in specific cases like antibiotic recovery or irritable bowel syndrome (IBS). So, in conclusion, not all probiotics are scams, but not all are worth your money either. Always read the labels, check for clinically studied strains, and don't underestimate the value of a fiber-rich, whole-food diet in supporting your natural gut flora.
FMT involves the transfer of processed stool from a healthy donor into the gastrointestinal tract of a recipient. It has achieved cure rates of >90% in recurrent Clostridioides difficile infections and is currently being explored for IBD, obesity, and even neurodevelopmental conditions. Challenges include donor screening, long-term safety, and standardization of delivery methods. The potential for engineered microbial consortia may one day replace crude FMTs with precision-targeted therapies.
The future of microbiome medicine, as with all areas of healthcare, lies in personalization. Advances in metagenomic sequencing, metabolomics, and artificial intelligence are enabling stratification of patients based on microbial profiles. Microbiome-derived biomarkers are being investigated for predicting drug response and disease risk. Bioengineered bacteria capable of delivering therapeutic payloads (e.g., anti-inflammatory cytokines or checkpoint inhibitors) are in preclinical development. As these therapies advance, they raise ethical considerations regarding microbial ownership, privacy, and equitable access. But this also forces us to ask more philosophical questions: if these microbes shape who we are, influence our health, and even our thoughts and emotions, do we truly own them, or are we simply their hosts?
Let’s bust some false rumours

Myth: “All probiotics are good for everyone.”
Fact: Probiotic efficacy is strain-specific and context-dependent. Some may worsen symptoms in people with small intestinal bacterial overgrowth (SIBO).

Myth: “More bacterial diversity is always better.”
Fact: Not necessarily. In some cases, higher microbial diversity can be linked to certain diseases because harmful bacteria thrive in environments with less competition and may take advantage of an imbalanced environment with more opportunities to grow.

Myth: “Diet changes take months to affect the microbiome.”
Fact: Shifts can occur within days, though long-term healthy dietary patterns are more stable.
The human gut microbiome is a master regulator of physiological health, from nutrient extraction to mood stabilization. As science deepens our understanding of these microbial networks, the potential for innovative interventions, from custom-tailored diets to engineered microbial therapies, is becoming a reality. However, personalized approaches, clinical trials, and clear regulatory guidelines will be essential in translating microbiome science into safe, effective medical practice. In the meantime, maintaining a fiber-rich diet, minimizing unnecessary antibiotics, and nurturing gut health remain the foundation of preventative medicine.
Monstrous Medical Mistakes
Imagine checking into a hospital for a routine tonsillectomy and waking up with one of your kidneys missing. No, this isn’t the plot of a new horror movie, it’s just another day in the crazy world of real-life medical errors.

Medical mistakes are no small slip-up. In fact, according to a Johns Hopkins study, they're the third leading cause of death in the United States, after heart disease and cancer. Estimates suggest that 250,000 to 440,000 US citizens die every year from preventable errors in healthcare. And for those who survive? Some walk out with surgical tools still inside them, the wrong limb amputated, or even memories of waking up in a morgue. Surgical items like sponges are left inside patients roughly 1,500 times a year, that’s four times a day!

I will deep dive into the world’s most jaw-dropping, and legally horrifying medical mistakes. This article is case-study heavy, filled with what we call ‘never events’. That is the official term for medical errors that are so serious and so preventable they should literally never happen. And yet… they do. Spoiler: they happen more than you think.

Let’s scrub in.
You’d think ‘left or right?’ would be the sort of thing double-checked before surgery. And yet, the US sees an estimated 40 to 60 wrong-site surgeries every single week. Imagine that on a global scale.

Take Willie King, a diabetic patient at University Community Hospital in Florida. In 1995, he was prepped for an amputation of a gangrenous leg. The operation went smoothly. Right about until someone realized they had amputated the wrong leg. Yes. The healthy one. King eventually received a $900,000 settlement and became a grim milestone in the history of medical malpractice.

Then there’s Rhode Island Hospital, where surgeons in 2007 operated on the wrong side of a patient’s brain not once, not twice, but three separate times in a single year. One of the patients died. The hospital was fined and subjected to intensive oversight, though no amount of paperwork could stitch back what was lost.

At this point, the public questions, what do they teach in medical school?
One of the most common ‘never events’ is the retention of surgical instruments inside patients. Up to 1 in every 5,500 surgeries results in a retained object. Yes, they close and sew people up with stuff still inside them. Most common are sponges, sometimes discovered years later when a patient complains of ‘weird stomach cramps’.

Consider the unnamed woman in the UK who spent six years suffering from abdominal pain after a routine operation. Eventually, doctors discovered a surgical sponge had been left inside her, and over time, it had fused into her intestines. Emergency surgery saved her, but barely.

Then there’s Ma Van Nhat, a Vietnamese man who complained of mysterious stomach pains, for 18 years straight. In 2017, doctors finally discovered the culprit: a 30-centimeter pair of forceps left behind during an operation in 1998. The man had been carrying them like a metallic parasite for nearly two decades. Did TSA ever ask why he was carrying surgical hardware internally?

And now for the showstopper: Dirk Schroeder, a German retiree who underwent prostate surgery in 2009. After the operation, he complained of severe pain. Doctors dismissed him, saying it was normal to feel moderate discomfort during recovery. However, his pain wasn’t tolerable and not until follow-up procedures, did the doctors reveal that 16 surgical objects had been left inside his body, including a needle, a six-inch roll of bandage, a swab, and several broken pieces of equipment. It took two additional surgeries to remove them all.

Hospitals often complain about equipment shortages, maybe that’s because half of them are still inside patients.
Roughly 1.5 million people in the US are harmed each year due to medication errors. Some of these mistakes are so bizarre you’d think I made them up for dramatic effect.

Let’s start with RaDonda Vaught, a nurse at Vanderbilt University Medical Center. In 2017, she was tasked with administering Versed, a mild sedative, to an elderly patient. Instead, she pulled Vecuronium, a paralytic agent, from an automated dispensing cabinet. The patient, Charlene Murphey, was left brain-dead and died shortly after. Vaught was eventually convicted of criminally negligent homicide igniting a global debate about accountability in medicine.

Swiftly shifting from this tragedy to complete absurdness. In 2012, Ilda Vitor Maciel, an 88-year-old woman hospitalized in Rio de Janeiro after suffering a stroke, was accidentally injected with chicken soup into her veins instead of into her feeding tube. The nurse, reportedly named Ana, mistakenly administered the soup intravenously. Maciel died shortly after the incident, and her family filed a medical malpractice lawsuit against the hospital.

In a case, which sounds like a textbook medical miscalculation, a 9-month-old girl at Children's National Medical Center in Washington DC, died in 2001 after a nurse administered a massive overdose of morphine due to a misplaced decimal point. The intended dose was 0.5 mg, but the infant received 5 mg, ten times the appropriate amount. Due to some miracle, the baby survived.

This is why medical students are taught ‘never trust your gut, trust the dosage sheet’.

The World Health Organization (WHO) estimates that medication errors cost approximately $42 billion USD annually, representing 1% of total global health expenditure. This substantial financial burden highlights the widespread impact of medication errors worldwide. Recognizing this, WHO launched the ‘Medication Without Harm’ initiative in 2017, aiming to reduce severe, avoidable, medication-related harm by 50% globally over five years.
Scalpel, Check. Anaesthesia, Check. Sanity? Missing..
Checklists save lives. Ego kills. Humility is the best medicine.
Surgery is already a stressful experience. Now imagine the operating table bursting into flames mid-procedure.

In one reported case, a 33-year-old pregnant woman underwent an elective Caesarean section under spinal anaesthesia. During the procedure, the surgical site caught fire due to the use of an alcohol-based antiseptic and electrocautery equipment. The fire resulted in 17% second-degree superficial partial-thickness burns, leading to circulatory shock. Fortunately, both mother and newborn were discharged without further complications.

Similar incidents have been reported across the US and UK. In one particularly gruesome case, a man suffered third-degree facial burns during a simple facial mole removal. The cause? An oxygen-rich environment combined with flammable prep-agents and electrocautery tools.
Misdiagnosis affects hundreds of millions of people globally each year, and one in three of those cases result in serious harm or death.

In 2020, Kayleigh Colegate, a 33-year-old mother of two from Shropshire, UK, visited the hospital multiple times complaining of shortness of breath, dizziness, and chest pain. She was repeatedly told it was a chest infection. In reality, it was a pulmonary embolism, and she died before receiving proper treatment.

In the US, a woman suffering chest pain was told it was just indigestion. She was given antacids and sent home. Hours later, she died of a heart attack.

Turns out Google might’ve done a better job, though it always thinks you have cancer. And as a chronic ‘Google-er’, I can vouch, I’ve misdiagnosed myself with terminal illnesses plenty a time. My advice is, see a trained professional, they have a lesser chance of getting it wrong.
There’s something especially terrifying about being declared dead when you’re still alive. I know, that’s a horror movie plot that would haunt my nightmares as a kid.

In July 2010, Maria de Jesus Arroyo, an 80-year-old grandmother from Los Angeles, was pronounced dead after suffering a heart attack. She was then routinely placed in the morgue freezer at White Memorial Medical Center. Days later, morticians found her body face down with a broken nose and facial lacerations. A pathologist concluded that she had been alive when placed in the freezer and died from asphyxiation and hypothermia while trying to escape.

Even more surreal, in 2014, Janina Kolkiewicz, a 91-year-old woman in Poland, was declared dead and likewise placed in a morgue freezer. Eleven hours later, staff noticed movement in her body bag. Upon rushing to get her out, she was determined to be alive and well, with no lasting health issues. I imagine she walked out of the morgue physically fine, but emotionally shaken for the rest of her life.

You know healthcare’s in trouble when the patients must call for their own resuscitation.

Some mistakes aren’t just about a single doctor or nurse, they’re due to institutional errors, when the entire system fails.

Between 2005 and 2009, Stafford Hospital in England saw as many as 1,200 unnecessary patient deaths, due to neglect, staffing shortages and systemic dysfunction. This complete collapse in patient care standards created what would become known as the Mid-Staffordshire NHS Trust scandal. It led to a public inquiry and major reforms in NHS safety regulations. The phrase ‘careless killing’ was used in the media, in association to this case.

In a separate case, Dani Marie Schofield, a nurse at Asante Rogue Regional Medical Center in Medford, Oregon, was accused of replacing patients' prescribed fentanyl with tap water in intravenous drips. This led to bacterial infections and at least one death. She caused mass under-medication of post-op patients for weeks before being caught. The hospital faced multiple lawsuits totalling over $477 million.

In these rigged systems, even good doctors get drowned in bad policy.
Medical mistakes are not just human errors; they are often systemic failures. From checklists to culture, every part of the healthcare system plays a role in patient safety. What I’ve discussed here are the worst-case scenarios, the stuff that turns a routine check-up into a Netflix series. But they also serve as a chilling reminder that in healthcare, attention to detail is everything. ‘Never-events’ mustn’t happen. The stakes are too high.

If you didn’t trust hospitals before, you’ve now got the receipts, but as horrifying as these mistakes are, they are the exception, not the rule. The vast majority of doctors and nurses are incredibly competent, compassionate, and cope well under the enormous pressure of the consequences behind their actions. Modern medicine saves far more lives than it harms, and thanks to improved safety protocols, electronic systems, and transparency, errors like these are becoming increasingly rare. So keep the faith, just maybe double-check that consent form.
Interviews with Experts
Lorem Ipsum is simply dummy text of the printing and typesetting industry. Lorem Ipsum has been the industry's standard dummy text ever since
Name
Information about the expert
What inspired your career path?
Text text text text text text text text 
text text text text text text text
Advice for students?
Text text text text text text text text text text text text text text text
Know someone who’d be a great interviewee?
Email Address
“After my knee surgery, the convenience of online consultations made my recovery smoother than I could have imagined.”
- Name
- Name
“Managing chronic conditions like diabetes requires a lot of vigilance, but the medicine refill system has simplified my life.”
Contact Number
“After my knee surgery, the convenience of online consultations made my recovery smoother than I could have imagined.”
- Name
Projects
The Lung Prize by Imperial Medical School
Research in a team of aspiring medics on various respiratory conditions, including Cystic Fibrosis, Idiopathic Pulmonary Fibrosis, Chronic Obstructive Pulmonary Disease, Long COVID and severe persistent Asthma. We're proposing an innovative treatment to such respiratory conditions, which all involve an overproduction of mucus, causing buildup in the lungs. Our solution, RespiraSense™, is a smart inhaler with mucolytic nanoparticles, a spacer, and visual/haptic cues. Lookout for more on this, once our submission is reviewed.
Neuroscience and Neuroimaging specialization from Johns Hopkins University
I completed a series of courses, from fundamentals of neuroscience in neuroimaging, to neurohacking in R. The latter few were extremely challenging, as it involved a lot of coding, and forced me to revise some of my programming knowledge from computer science GCSE and other experiences in coding. I now feel very confident in principles of fMRI, and have some understanding of R coding, in addition to my previous Python, C++ and C#.
IB Extended Essay
I am writing my IB Extended Essay on the effectiveness of cancer treatments in economically varying healthcare systems in Brazil (private vs. public SUS). Alongside my independent research on the subject, I will be attending a Pre-College Program at Harvard University on 'Molecular Oncology: Unravelling Molecular Biology Techniques'. Through this, I hope to gain a deeper understanding of the biological processes involved in battling cancer, and will attempt to delve into additional research in the field with the leading professor of the program.
AI in Healthcare specialization from Stanford University
2-month internship at VR Inclusion, a med tech startup dedicated to developing software that provides crucial support for individuals requiring assistance in developing social, behavioural, communication, and life skills, with a special focus on autism. My responsibilities encompassed assisting in conducting in-depth research related to autism and the technologies used to support these individuals, compiling valuable vendor information, preparing and delivering presentations to attract investors, and assisting with translations to ensure that our research and materials were accessible to a wider audience.
Summer Internship at VrInclusion
I visit the senior residents weekly to connect with them through games and conversation, helping reduce loneliness and cognitive decline. Research shows how intellectual games and consistent communication can offset lots of complications that are common among the elderly, such as Alzeihmer's and chronic loneliness. As a team of volunteers, we aim to combat social exclusion, challenge ageism, and foster intergenerational empathy by amplyfying older voices.
Volunteering at a nursing home in Cheltenham (Astell House)
Successfully completed 5 college courses, which spanned across units about the structure of the healthcare system, including different intermediaries involved; the vastness of clinical data used in medicine; evaluations of how machine learning and AI can be applied to healthcare professions; and a self-led project at the end. This project opened my eyes to the amount of processing that clinical data goes through, in order to simplify it for doctors to view and make diagnoses from.
Tutoring services
Service
Offering: KS3/GCSE Bio/Chem/Maths (group or 1:1)
We’re also more than happy to message you about any academic concerns or join a quick Zoom call for study tips, and general advice!

Please contact me if you’re interested and make sure to mention times that would be suitable for you (if applicable), so that I can best allocate me or one of my team to help you!

Remember you’re welcome to send an email with any direct questions, I will respond promptly.
Hello! Are you struggling in a particular class or can’t get the hang of a topic? Maybe you would benefit from a sixth-form student, who can relate to the pain, helping you grasp the concept. Me and my team of volunteers are happy to offer 1:1 tutoring in biology, chemistry, physics and math at a level of KS3 / iGCSE / GCSE (including add math and further math qualifications).
Booking
arina@kozerog.com
Reviews
Daniela
Student
Arina has helped me grasp concepts in biology that I’ve never been able to understand and was very patient with me. Highly recommend!
Melanie
Student
I had tutoring for iGCSE chemistry and what I enjoyed most was the unique way she presented topics. She helped me remember topics by associating it with other things, using acronyms, or helpful diagrams. Thank you!
Artem
Student
Since I started having lessons here, I’ve jumped 2 grades in maths. Something that I feel helped was doing a past paper in my own time, and then debriefing the questions I couldn’t answer during our lessons. Very helpful.
Nestia
Student
Arina has helped me grasp concepts in biology that I’ve never been able to understand and was very patient with me. Highly recommend!
Alex
Student
Chemistry has always been my worst subject, so I asked Arina to help me prepare for my GCSE exam. Just a few sessions made me really confident in the organics unit, which I had found particularly challenging.
Name
Student
/
Daniela
Student
Arina has helped me grasp concepts in biology that I’ve never been able to understand and was very patient with me. Highly recommend!
Melanie
Student
I had tutoring for iGCSE chemistry and what I enjoyed most was the unique way she presented topics. She helped me remember topics by associating it with other things, using acronyms, or helpful diagrams. Thank you!
Artem
Student
Since I started having lessons here, I’ve jumped 2 grades in maths. Something that I feel helped was doing a past paper in my own time, and then debriefing the questions I couldn’t answer during our lessons. Very helpful.
Nestia
Student
Arina has helped me grasp concepts in biology that I’ve never been able to understand and was very patient with me. Highly recommend!
Alex
Student
Chemistry has always been my worst subject, so I asked Arina to help me prepare for my GCSE exam. Just a few sessions made me really confident in the organics unit, which I had found particularly challenging.
Name
Student
/

Want to contribute an article?
LinkedIn
Email me at arina@kozerog.com with your idea!
E-mail
Contact