Health

Clinical Trials That Went Horribly Wrong

French Biotrial Tragedy In early 2016, the French company Biotrial recruited 128 healthy volunteers to participate in a clinical trial for a new drug. Under small… Trista - December 15, 2022

Clinical trials are a crucial step in getting FDA approval for any drug. Without them, how would we know the various side effects or if they are even effective? Most of the time, clinical trials are successful, with few negative outcomes, and the drug receives its approval for use. But then there are some clinical trials that go horribly wrong and people paid for that with their lives.

Dan Markingson went to UM Fairview in 2003. MHealthFairview.org

The University of Minnesota Seroquel Experiment

It may be surprising to find out that universities can conduct clinical trials, but they can take place in many locations. FDA (the Food and Drug Administration) requires five steps for drug clearance. These are Discovery and Development, Pre-clinical Research, Clinical Research, FDA Drug Review, and FDA Post-Market Drug Safety Marketing. No clinical trial can begin without the proper authorizations. The first set of approvals needed would be from governing bodies, and that’s only after showing positive results in laboratory tests and animal studies to test the therapy’s safety and efficacy. The other part of authorization involves the informed consent of the human participants, and that’s where this particular experiment went off the rails.

Dan Markingson and his mother, Mary Weiss. Mother Jones

The Death of Dan Markingson

In 2003, Dan Markingson was experiencing delusions, and he was diagnosed with schizophrenia and admitted to the University of Minnesota Medical Center. There, they quickly accepted him into a clinical trial testing three different medications for his diagnosis: Seroquel, Risperdal, and Zyprexa. His delusions immediately worsened, and his mother responded by trying to remove him from the program. The administration threatened to have him committed to a mental facility if he dropped out of the trial until she learned their real motivation: his participation earned the school $15,000.

Dan Markingson. A Family Photo

Protect Your Mental Health

Trapped in a program that caused worsening delusions, Dan’s mental health deteriorated until he eventually took his own life. He left a note that read: “I went through this experience smiling!” It later came to light that a judge ordered Dan to go into the UM Medical Center on the advice of Dr. Stephen Olson – a UMN Doctor – but only if he followed Dr. Olson’s treatment plan. His plan? Enrolling in the clinical trial. Dr. Olson then delegated the responsibility for Dan’s treatment to a social worker with no medical training and wholly ignored his worsening condition and his mother’s pleas for help, despite his assessment just days prior that Dan, in Dr. Olson’s own words: “lack[ed] the capacity to make decisions regarding [his] treatment.”

The death of Dan was only the beginning. The Guardian

The Unfortunate Aftermath

The worst part is that his mother sued the school after Dan’s death, but they refused to take responsibility for their actions. Unfortunately, Dan was not the only one in the trial to take his own life. Four others attempted suicide, and one of them succeeded. The university hid behind statutory immunity and instead sought to make Mary pay $56,000 for his treatment! It then used those costs as leverage to make her abandon her suit against them. Not everyone at UM is an evil person, though. A group of bioethicists called on the university’s Board of Regents to conduct an independent investigation into the clinical trial. Unsurprisingly, their lawyer determined that no follow-up was necessary.

One of six men hospitalized after the clinical trial, Stephane Schubhan still suffers from neurological damage. NRP

French Biotrial Tragedy

In early 2016, the French company Biotrial recruited 128 healthy volunteers to participate in a clinical trial for a new drug. Under small doses, none of the participants experienced any side effects. Once the quantities increased, six of the participants became very sick, which ultimately led to the death of one of them, with permanent damage to the rest. It was later discovered that the drug was known to have similar effects on dogs during pre-clinical trials, yet it moved to human testing.

French Health Minister Marisol Touraine left, and Professor Gilles Edan, the chief neuroscientist at Rennes Hospital. Forbes

The Death of Guillaume Molinet

After five days of taking the experimental drug that might help dull his nerve pain, Guillaume Molinet checked into the hospital. An MRI scan showed a massive stroke in the brain stem. He was declared brain dead that day, and a week later, he passed away. Five other volunteers on the highest dose of the clinical trial drug that checked into the hospital, with four exhibiting similar symptoms and showing identical brain injuries in scans. Thankfully, none of the other participants at lower doses experienced any problems, though that seemed to be more of a stroke of luck than by design.

The drug worked on the endocannabinoid system in the brain as well as other parts. Shutterstock

What Went Wrong with the Biotrial Case

The people running the trial appeared to lack any logic or common sense. They gave extremely high doses of the drug: almost 40 times more than needed to subdue any pain. They’d also completely ignored the fact that the drug had not really even shown indications of treating pain in the first place and moved it to human trials anyway. Despite all these missteps, everything they did was legal, but not necessarily intelligent or wise. The drug was intended to work on the endocannabinoid system in the brain. It is likely that it also worked on unintended parts in the brain which prevented an enzyme called fatty acid amide hydrolase, or FAAH, from breaking down a molecule in the brain that helps with our response to pain and anxiety.

German drug company Grunenthal remained silent about its drug for fifty years before issuing an apology. Smithsonian

The Thalidomide Trials

Thalidomide was initially marketed in West Germany in 1957 as an over the counter drug for anxiety, sleeplessness, tension, and morning sickness. We may recognize the name now because of its adverse effects on pregnancy. Nevertheless, at the time, they thought it was a safe drug. Researchers found it to be a non-sedative with no risk of a hangover or dependency. They released it to market after testing with no need for a prescription. Most importantly, they perfromed zero tests for teratogenic potential, meaning anything that could cause fetal abnormalities.

They didn’t know this drug would harm their unborn children. RX

Not for Pregnant Women

In 1961, Dr. William McBride and Dr. Widukind Lenze observed a relationship between thalidomide and congenital malformations like limb and bone abnormalities and gallbladder abnormalities. The drug was pulled from most markets that year and banned from all markets by 1970. By then, it had affected an estimated 10,000 infants worldwide, not including those stillborn or miscarried during pregnancy. Thalidomide’s main hazards and side effects, not including death, include fetal impairment like bilateral limb atrophy, bilateral limb absence, missing fingers and toes, malformation of the digestive tube, malformation of the duodenum, malformation or absence of the anus, and vital organs injury.

Survivors of the horrible trial. The Austrialian

Saving the Babies

The FDA did not approve Thalidomide in the 1950s and 1960s due to the efforts of one Dr. Frances Oldham Kelsey. She refused to approve the application because of what she felt was inadequate evidence of the drug’s safety, despite its use worldwide. She insisted on scientifically reliable evidence, which she felt the application lacked. Just a year later, Dr. McBride and Dr. Lenze made their discovery, and Dr. Kelsey was awarded the highest recognition attainable for a U.S. civil servant for her role in saving thousands from death or life-long incapacitation. In October 2000, Dr. Kelsey was also inducted into the National Women’s Hall of Fame. In 1998, Thalidomide was approved in the United States as a treatment for cancer and is on the World Health Organization’s List of Essential Medicines, and is also available as a generic medicine. However, it should not be used by anyone trying to conceive a child or by anyone breastfeeding or pregnant.

Jesse Gelsinger was only two years old when he was diagnosed with Ornithine Transcarbamylase Deficiency Syndrome (OTCD). AP Photo/Family Photo via The Arizona Daily Star

Gene Therapy Clinical Trial

Babies born with Ornithine Transcarbamylase Deficiency Syndrome (OTCD) usually don’t reach adulthood, but Jesse had a milder version of the deficiency. OTCD is a rare metabolic disorder that builds ammonia up to lethal levels in the blood. Though he was able to manage it with a low-protein diet and a regimen of anywhere between 30 – 50 pills every day, he was incredibly interested to hear more about the trial that could treat it. Researchers at the University of Pennsylvania were developing a clinical trial to inject patients with a working version of the gene he didn’t have – attached to a cold virus to make it more spreadable.

Jesse’s story and the failed gene therapy made national news. CBS News.

What Happened to Jesse? (Part 1)

Jesse was the 18th person to receive the injection. Previous patients had experienced flu-like symptoms, but he reacted much more severely. He then showed signs of jaundice, blood clotting, and organ failure. Four days later, he was declared brain-dead and taken off life support. After a thorough investigation, many troubling things came to light. The researchers had not retooled their eligibility criteria despite promising the FDA that they would.

Clinical Trials

What Happened to Jesse? (Part 2)

Two patients experienced severe side effects, but it was not reported, nor was the study put on hold. Jesse’s pretrial test results indicated that his liver function was poor enough that he should not have received the drug at all – worse yet, Jesse was not informed of any of these issues, so he did not have informed consent. Jesse’s family sued, and the University settled for an undisclosed amount. In January 2000, the FDA suspended human research at UPenn’s Institute for Human Gene Therapy, and the University eventually shut the program down.

Potti is a former Duke University associate professor and cancer researcher. Biz Journals

Anil Potti’s “Miracle” Cancer Drug

Anil Potti is the scariest kind of doctor. He is the one who promises you the world when he knows he is lying. He promised cancer treatments with an 80 percent cure rate and fabricated data that convinced medical professionals that he could save 10,000 lives a year. Anil Potti graduated from Christian Medical College in India in 1995, then completed an internship in Internal Medicine at the University of North Dakota School of Medicine in 1999. He moved on to complete training in hematology and oncology at Duke University in 2006. However, he resigned in 2010 after discovering flaws in his research and allegations of embellishments in his resume.

There is not a miracle pill for cancer treatment. Shutterstock

Lies and Deceit

Potti and his team were accused of falsifying data regarding personalized cancer treatments. However, scientific journals published these findings. As of 2020, journals retracted 11 of his research publications. Furthermore, one has received an expression of concern, and two others have been corrected. Potti’s work was funded through the National Institutes of Health (NIH), the National Cancer Institute, the Howard Hughes Medical Institute (HHMI), and others. Joyce Shoffner was one of Potti’s victims. She signed up to participate in his cancer-curing clinical trial, undergoing a painful breast biopsy and following a regimen of Adriamycin-Cytoxan (AC) chemotherapy, which caused her blood clots and diabetes as well as PTSD from the trial. Keep reading for facts about clinical trials that won’t horribly wrong.

Macular degeneration is the leading cause of vision in people 50 years and older. Shutterstock

Stem Cell Vision Treatment

In any clinical study, researchers treat patients as gracious volunteers that are helping further science. Sometimes, there’s a financial compensation aspect to it too. One thing is for certain: they never ask trial participants for payment in exchange for treatment. At least, that’s how things are supposed to work. In this unfortunate stem cell vision treatment, they charged three women $5,000 for their trial. Sadly, they ultimately lost most, if not all, of their eyesight.

In January 2017, a study asking for elderly participants with macular degeneration offered stem cell therapy to treat their eyesight. All three women reported bleeding and retinal detachment just days after their treatment. One woman was completely blinded, while the other two lost most of their sight. The researchers have tried to erase the trial’s existence; when you visit the records of it online, it says that the study was “withdrawn before enrollment,” which is a lie.

Do you support science for adult stem cells? Healthy MD

What Happened to the Patients?

The patients had fat cells removed from their abdomens and a standard blood draw for lab work. The fat tissue from their cores was processed with enzymes to obtain stem cells, then platelet-dense plasma was isolated from the blood. Doctors mixed the cells with the plasma and injected it into their eyes. The process took less than an hour. However, they treated both eyes at the same time. That is another red flag, as typically a clinical trial would wait to see how one eye responds to treatment before exposing the other. Though the patients had no way of knowing this, the trial had no real scientific basis. It had no hypothesis based on laboratory experiments, no control and treatment groups, no collection of data, no clear patient masking (to see who was and was not getting treatment), and no plans for follow-up.

Manufactured (CAR)-T cells ready for infusion into a patient. Cancer.gov

Hopeful for a New Leukemia Treatment

Researchers are always searching for a cure for cancer. Leukemia is a particularly terrifying type of cancer. So Juno Therapeutics’ clinical trial tested their new cellular-level treatment that would attack malignant cells until they appeared to vanish, it came out. No wonder researchers in the cancer community were calling it the “fifth pillar” of cancer treatment. The treatment, chimeric antigen receptor (CAR)-T cell therapy, or CAR-T for short, was not specific to Juno Therapeutics’, but its results would certainly affect the entire scientific community.

Different kinds of leukemia can require different types of treatment. Hematology

Leukemia CAR-T Trial Fails

The study in July 2016 caused the death of three leukemia patients from cerebral edema, or swelling in the brain. Representatives from the company admit that this is common in patients receiving CAR-T treatments, immune system reactions, and increased neurological toxicity. After the news of the deaths, the company’s stock fell by 27 percent and their practices went under FDA review. The researchers did not know how to properly gauge patient immune cells’ heterogeneity. Plus, once they injected the patient, something else happened. The rapid multiplication of the CAR-T cells led to neurotoxicity. Juno learned from its mistakes and started monitoring T-cell subtypes in other CAR-T trials. Keep reading for facts about clinical trials that won’t horribly wrong.

Hoiyan Nicole Wan was barely 20 years old when she died at the University of Rochester. Henry Ford

New York Lidocaine Disaster

Like any college student, Hoiyan Nicole Wan was in search of a little extra pocket money. She heard of a clinical trial that paid $150 for a bronchoscopy to see the effects of pollution on her respiratory system, so she signed up. Wan had not planned on the researchers taking more cell samples than initially proposed, and increasing the anesthetic, Lidocaine, by about four times the maximum dosage. The college student was released feeling very weak and in pain, and two days later she was found dead.

A tragic ending. National Geographic

What Went Wrong?

Her autopsy revealed that the Lidocaine levels she received were lethal, causing her heart to stop beating and her organs to shut down. She had a heart attack the day of the study, in fact, after initial struggles with breathing and then going to the emergency room. Her parents settled with the university. They did not release the full details. However, it does call for a full scholarship for a Chinese-American student for eight years, an annual lecture on the ethical and safety issues in medical research involving humans, and a memorial in Ms. Wan’s name. Keep reading for facts about clinical trials that won’t horribly wrong.

Ellen Roche worked at Johns Hopkins, the very place that led the trial that killed her. JH Medicine

John Hopkins Asthma Trial

Ellen Roche was a 24 year old technician at Johns Hopkins Asthma and Allergy Center, where she heard of a trial recruiting healthy volunteers to help doctors discover the reflex that protects the lungs of healthy people against asthma attacks. The researchers would provoke a mild asthma attack by having her inhale hexamethonium, a medication used for treating high blood pressure in the 1950s and 1960s. She signed up for the study and quickly developed a cough, though the researcher administering the trial chalked it up to a common cold. Her condition worsened over the next week. Doctors put her on a ventilator. Ellen Roche’s lung tissue dissolved and her blood pressure plummeted. Then, her kidneys began to fail. She died on June 2, 2001, a month after entering the study.

It can take months or even years to hear an apology — if ever. JH Medicine

Admitting Fault

Medical officers from the trial now admit that the hexamethonium was either solely responsible or played a contributory role to her illness, but that doesn’t make accepting her death any better. In fact, to make things worse, participants learned the FDA did not even approve of the drug after the trial. In their consent forms, they merely referred to it as a “medication” instead of clearly indicating that it was experimental or listing the risks. Keep reading for facts about clinical trials that won’t horribly wrong.

Approximately 95% of the animals used for scientific research in the U.S. are rodents. Standford Med

What Works on Animals May Not Work on Humans

Since chimpanzees share more than 99% of DNA with humans, and mice share more than 98%, they are susceptible to many similar health problems as humans like cancer, diabetes, and heart disease. Researchers are legally and morally obligated to ensure the health and well-being of the animals in their care. They never set out to harm them; animal testing is essential in drug approval. Animal trials have helped scientists learn about disease stages and how to treat or even cure them. Currently, animal use is even a mandated part of drug development. Current federal laws in the U.S. require proof of safety and effectiveness in animal trials before they move into human trials. In our last entry, we’ll look at a clinical trial where successful animal testing didn’t translate to safe human testing.

Potential wonder cure TGN1412 was not so wondrous. Patients had extreme pain before leading to organ failure. The Guardian

Theralizumab – AKA The Elephant Man Trial

Theralizuman is also as TGN1412, CD28-SuperMAB, and TAB08. Thomas Hunig developed it at the University of Wurzburg in 2006. The purpose of the drug was originally to treat B cell chronic lymphocytic leukemia (B-CLL) and rheumatoid arthritis by binding to receptors on the T-cell. The six human participants were assumed that the worst symptoms they’d experience were likely a headache and nausea, but the trial soon spiraled into one of the most infamous medical trials in British history. Shortly after receiving their doses, all patients began experiencing severe pain and vomiting uncontrollably. One patient lost his fingers and toes, another had to have this foot partially amputated. The trial is “the Elephant Man Trial” because one patient’s head swelled up so much that his girlfriend teased him about looking like an elephant.

Severe swelling occurred as a result. Penn Vet

The Cytokine Storm Symptoms

Doctors now know to call the symptoms men experienced cytokine storm. It is where their temperatures soared, their organs failed, and some of their bodies swelled past recognition. One of the participants, Rob Oldfield, had fluid seeping into his lungs and had to breathe through a mask. Ryan Wilson spent four months in the hospital and battled pneumonia, septicaemia and dry gangrene. His father had warned him the night before the trial: “Don’t do it. Your body is a temple.” The volunteers still suffer from weakened immune systems and other side effects.

Participant Rob Oldfield, 21 at the time, had amputated all of his toes and parts of his fingers. Huffington Post

What Went Wrong with the Elephant Man Trial?

Though knowing what went wrong with the trial is difficult, there are a few theories about why it was so ill-fated. One theory claims that the period of drug injection was to blame. Researchers spent over an hour and a half injecting the drug carefully into the animals. However, injecting the entire dosage into the human participants took a mere six minutes. Another theory suggests that animal testing did not translate to human testing accurately because it was done on a macaque instead of a bonobo, so the animal-to-human DNA match was only 94% instead of 98%.

What’s good for the goose is not always good for the gander. Pharma

Parexel on Animals vs Humans

Oddly, one source said that the first infusion in six patients was five hundred times smaller than that found safe in the animal studies, but the volunteers still suffered life-threatening organ failure. Clearly, a disconnect between animal and human studies happened here, resulting in a hugely unfortunate incident for those involved. Parexel, the drug company that ran the clinic where the trial was carried out, was found to have acted within all proper protocols. However, a final report did indicate that the trial had not adequately considered the safe dosage for humans. The company that developed the drug, TeGenero Immuno Therapeutics went bankrupt later that year.

Advertisement