Posts Tagged ‘body’

Is it possible to come back from the dead? Australia’s first body-freezing facility explores the boundaries of mortality – Neos Kosmos

Freezing your body after death with the hope of coming back to life one day sounds like something out of a science fiction movie.

Southern Cryonics, the first body-freezing facility in the Southern Hemisphere, tries to turn this idea into reality.

Were sort of in a race against time, says Southern Cryonics director, Peter Tsolakides, to Neos Kosmos.

Cryonics, coming from the Greek word kros for icy cold, involves the preservation of legally declared dead bodies at extremely low temperatures for potential future revival.

The facility in Holbrook, New South Wales, uses this practice, with the expectation that one day, advancements in medical technology and science will restore patients to health and in the young body.

But the timeframe of this future remains uncertain.

Tsolakides says once you preserve a body, you can keep it (stored) for thousands of years, but the chance of coming back depends on when you freeze it.

A matter of life after death

He says, currently 50 people, are willing to take the risk for a chance at life after death, and the number is growing.

This group consists of 35 investors each contributing $50,000 to $70,000, and 15 subscribers or customers who have paid $150,000 through life insurance.

Tsolakides says there are no guarantees, despite how great the dream of being brought back from the dead might be for some people.

Most of them know something about it (cryonics), but they also look at it and say, look, theres no guarantees, but theres a chance.

And that chance versus being buried in the ground or cremated is a much higher chance coming back.

He estimates the chance of a well-preserved body being revived in 200 years to be around 20%.

He says although its hard to predict what the world will be like in be like in 1,000 years, bodies might be revived when technological advancements have found the key to immortality.

In the real world, nobody will be dying, and most diseases will be cured. So, we will know how to prevent death in a sense, and the next step is to bring back those who have already died, but in a good condition.

Tsolakides says that while they dont know how to bring a person back to life, current developments give you inklings, of what the future is going to be like.

He says progress has to start somewhere, and right now billions are invested in medical research aimed at disease cures.

This includes groups working on brain revival, organ regeneration, cloning, and advancements in artificial intelligence and nanotechnology.

How does cryonics work?

When a person is declared legally dead at the hospital, a cooling process begins.

Chemicals are used to stabilise the body, lowering it to about ice temperature.

Once taken to the funeral home, the body is further cooled and infused with an antifreeze substance until it reaches about -80C.

Next, it goes to the cryonics facility, gradually cooled to -180C and preserved below that temperature, in a large vacuum flask container filled with liquid nitrogen.

Southern Cryonics Greek-Australian director says, theres a brief window of a few hours after legal death, where no deterioration occurs to the body.

Once preserved in liquid nitrogen, it can be stored for thousands of years due to almost no chemical or biological activity at that temperature.

Its a race against time to keep the temperature going down, he says.

But is it possible to freeze a human brain to revive it later?

If you catch them (bodies) under our optimal time, very little damage is occurring to the brain, but that doesnt mean that 200 years from now, that damage cant be repaired, Tsolakides says.

The facility can currently hold up to 40 patients, with each container fitting 4 bodies, but can expand to hold up to six or seven hundred patients if necessary.

The birth and evolution of Southern Cryonics

Tsolakides got interested in cryonics from a young age.

When he came back to Australia around 2012 after working overseas, he saw there were only cryonics facilities in the US and Russia.

He connected with like-minded people and talked about building one in Australia.

We started getting what we call founding members, says Tsolakides, each contributing $50,000 to kickstart the project, eventually totalling 35 members of a non-profit organisation.

We started the facility and that was how it sort of developed.

He says, they chose Holbrook, a small town with about 1,500 people, for a few reasons.

Land there wasnt expensive, and it was halfway between Melbourne and Sydney, making it accessible to over half of Australias population.

Holbrooks nearby Albury airport is crucial for quick patient transportation, and the support from the local council made the decision easier.

Another advantage is its proximity to liquid nitrogen suppliers along the Hume Highway, crucial for the facility.

Holbrooks low history of natural disasters made it a safe choice after a thorough analysis of several years.

The legalities

Tsolakides says Southern Cryonics got all the official approvals from the NSW Department of Health and the local council, to operate as a cemetery but uses a recognised funeral home for mortuary work.

The government groups that we work with helped us a lot. It wasnt like we got resistance or anything like that.

A good idea but not for everyone

Tsolakides was born in Israel to Greek parents.

His mother was from the Greek island of Syros, and his father from Athens.

They briefly lived in Greece before moving to Australia in 1955 when Tsolakides was five years old.

He has a degree in Chemistry and later pursued one in Business Administration.

Throughout his career, he worked primarily in marketing for an oil company.

He grew up and lived in Melbourne for many years before moving to Sydney, a place he now calls home.

His passion in cryonics sparked at about18 after reading Robert Ettingers book, The Prospect of Immortality.

At that age, he didnt worry much about death.

He assumed this will be everywhere, by the time he got old, but soon realised that very few people worldwide were interested in it.

He says that while some are intrigued by cryonics, most view it as a good idea but not for themselves.

Even the US organisation have about five to six thousand members only, with 400 or 500 people suspended, and theyve been going for 50 years.

But that didnt stop him for pursuing his curiosity around cryonics.

Keeping an eye on scientific developments

Tsolakides is determined to improve their techniques and increase success chances, despite challenges or doubts about cryonics.

He says Southern Cryonics along with overseas organisations is monitoring the best way to store a body, leaving the revival work to other scientists.

Its (suspending the body) physically possible to do it now, he says, but of course, you can always improve the processes.

While cryonics remains a controversial field and the chances of revival seem low now, it is yet to be seen whether future technology will ever be able to bring the dead back to life.

Read this article:
Is it possible to come back from the dead? Australia's first body-freezing facility explores the boundaries of mortality - Neos Kosmos

Cell and Gene Therapy Market Size to Reach USD 97.33 Bn by 2033 – BioSpace

According to the latest research by nova one advisor, the global cell and gene therapy market size was valued at USD 18.13 billion in 2023 and is anticipated to reach around USD 97.33 billion by 2033, growing at a CAGR of 18.3% from 2024 to 2033.

Request PDF Sample Copy of Report: (Including Full TOC, List of Tables & Figures, Chart)@ https://www.novaoneadvisor.com/report/sample/7720

The cell and gene therapy market provides therapeutic solutions related to genes and cells. The market deals with research & development, testing, production, and distribution of products and treatment procedures related to genes and cells. Hospitals, research laboratories, pharmaceutical companies, pharmacies, research institutions, and universities are involved in delivering the applications associated with gene and cell therapies. Gene and cell therapies are developed to prevent, treat, or potentially cure numerous diseases. The potential of these therapies to cure, treat, or prevent diseases that are life-threatening increases the demand and boosts the growth of the market. Gene and cell therapies are used in blood stem cell transplantation, gene editing, engineering of the immune system, tissue regeneration, in-vivo gene transfer, cancer treatment, and treatment of different disorders. These therapies can provide better results and enhance quality of life.

North America dominated the cell and gene therapy market in 2023. North America is a developed region that has developed healthcare and research infrastructure, better facilities, and government support that boosts the growth of the market. Governments in the North American region have a huge national budget for healthcare and research. Countries like the U.S. and Canada contribute to the growth of the market in the North American region. As of now, the FDA has approved 37 products for gene and cell therapy. The U.S. has the American Society of Gene & Cell Therapy (ASGCT) for professionals, scientists, physicians, and patient advocates that help advance knowledge, education, and awareness for discovering and developing clinical applications of gene and cell therapy.

The Canadian government is also focusing on improving health with the help of genes and therapies and is launching various programs to help with this. The government launched Disruptive Technology Solutions, which will help tackle the challenges associated with gene and cell therapies. The treatment procedures will be done to cure rare genetic disorders and chronic diseases.

Key Takeaways:

Immediate Delivery Available, Get Full Access@

https://www.novaoneadvisor.com/report/checkout/7720

The cell and gene therapy market is exploding globally

Ground-breaking developments in next-generation cell and gene therapies (CGTs) offer curative value for patients with few to no other therapeutic interventions for either maintenance or cure within specific disease areas, many of which include rare and ultra-rare diseases.The largest therapeutic area is cancer, followed by musculoskeletal diseases and eye diseases.Multiple approved products have been launched in global markets and the number of clinical trials continues to grow. In Europe, these therapies are classified under Advanced Therapeutic Medicinal Products (ATMPs) and are driven by a diverse set of scientific advancements including CAR-T, TCR-T, stem cells, siRNA, oligonucleotides, gene editing (CRISPR, Zinc Fingers, TALENs) and viral transfection.

The global CGT market is projected to grow at a compound annual growth rate of over 36 percent from 2019-2025, to ~ 10 billion. With more than 900 companies globally focusing on CGTS and over 1,000 clinical trials being conducted, the industry could see numerous approvalsas many as 10 to 20 new advanced therapies per year starting in 2025. Moreover, 33% of these clinical trials is being conducted in Europe.1

Global biopharma companies as well as smaller, venture backed-up start-ups are rapidly investing in this complex space. In 2018, about $13 billion has been invested globally in advanced therapies such as cell, gene and gene modifying therapies. In 2019, 19 CGT-related M&A deals worth over $156 billion were completed.

As with any innovative and disruptive technology, CGT developers face challenges along several key stages of the product life cycle. Compared to chemical-based pharmaceuticals, key success factors such as enabling patient access, managing supply chain and manufacturing operations, evidencing compliance with increasingly complex regulatory requirements and alternate business models impose a greater burden.

Segments Insights:

By Therapy Insights

The market for cell and gene treatments consists of companies (organizations, sole proprietors, and partnerships) that sell the therapies they have developed. Cell therapy is the transfer of whole, living cells derived from allogeneic or autologous sources, while gene therapy is the introduction, deletion, or alteration of the genome to treat disease. The market is made up of the money that businesses creating goods for cell and gene therapy make from the sales of those items.

Cell treatment and gene therapy are the two primary product categories in this field. Gene therapy is a field of medicine that focuses on altering cells' genetic make-up to treat disease or reverse it by repairing or replacing genetic material that has been damaged. Oncology, dermatological, musculoskeletal, and other applications are among the many that are used in hospitals, ambulatory surgery centers, cancer treatment facilities, wound care facilities, and other industries.

Cell & Gene Therapy Market Revenue, By Therapy Type, 2022-2032 (USD Million)

By Therapy Type

2022

2023

2027

2031

2032

Cell Therapy

13,396.01

15,621.48

29,433.95

57,138.21

67,757.69

Gene Therapy

2,067.97

2,502.14

5,406.11

11,864.27

14,480.51

By Therapeutic class

Based on application, the market is divided into cardiovascular disease, cancer, genetic disorder, rare diseases, oncology, hematology, ophthalmology, infectious disease, neurological disorders. Among these, the infectious disease segment dominates the market in 2023. The oncological disorder segment held a revenue share of 13.53% in 2023. Research and treatment in the biomedical domains of cell therapy and gene therapy. Both treatments have the ability to lessen the underlying cause of hereditary disorders and acquired diseases. Both therapies aim to treat, prevent, or perhaps cure diseases. By repairing or changing specific cell types, or by employing cells to transport a medication across the body, cell therapy tries to treat diseases. Cell therapy involves growing or modifying cells outside of the body before injecting them into the patient. The cells may come from a donor (allogeneic cells) or the patient (autologous cells)6. By replacing, deactivating, or introducing genes into cells, either inside the body (in vivo) or outside the body, gene therapy seeks to treat disorders (ex vivo).

The market for genetic disorders is expanding as a result of factors like the high prevalence of genetic and chronic disease cases and the growing government initiatives to raise public knowledge of genetic testing and diagnosis. Researchers are developing novel techniques for screening, diagnosing, and treating patients for a variety of cardiac diseases as they investigate the genetic roots of heart and vascular illness. Some researchers are looking for new ways to nine patients who are at risk for sudden cardiac death. Others are examining how medicines that could postpone or obviate the need for cardiac surgery could benefit patients with uncommon illnesses.

The intricacy of mitochondrial genetics and the diverse clinical and biochemical symptoms of primary mitochondrial disorders (PMDs) have shown to be a significant obstacle to the development of effective disease-modifying medications. A successful clinical transition of genetic medicines for PMDs is possible, according to encouraging evidence from gene therapy trials in patients with Leber hereditary optic neuropathy and improvements in DNA editing tools.

Cell & Gene Therapy Market Revenue, By Therapeutic Class, 2022-2032 (USD Million)

By Therapeutic Class

2022

2023

2027

2031

2032

Cardiovascular Disease

744.36

882.84

1,780.08

3,697.84

4,460.03

Genetic Disorder

1,643.41

1,922.21

3,665.70

7,202.20

8,566.52

Oncology

1,936.87

2,272.26

4,385.58

8,720.66

10,403.81

Hematology

1,196.56

1,396.75

2,642.34

5,150.06

6,113.36

Ophthalmology

835.60

972.46

1,817.62

3,500.15

4,142.33

Infectious Disease

4,420.18

5,206.30

10,210.05

20,628.98

24,708.86

Neurological Disorders

658.61

777.29

1,536.51

3,129.23

3,755.58

Others

4,028.39

4,693.50

8,802.17

16,973.35

20,087.70

By Delivery Method

The market is split into In Vivo therapy and Ex Vivo therapy according to the type of therapy. In vivo therapy market is anticipated to grow exponentially throughout the projected period. When it comes to gene therapy, there are two different methods: ex vivo and in vivo, setting aside cell therapies. The altered human gene must first enter the diseased person's cells for gene therapy to take effect. There are two methods for doing this; Genetic material is supplied in vivo to afflicted cells (cancer cells or other cells) that are still present within an individual's body. After cells are collected and exposed to the genome in Ex vivo, altered genes are transferred to a person's body.

See the original post:
Cell and Gene Therapy Market Size to Reach USD 97.33 Bn by 2033 - BioSpace

Why the sci-fi dream of cryonics never died – MIT Technology Review

The environment was something of a shift for Drake, who had spent the previous seven years as the medical response director of the Alcor Life Extension Foundation. Though it was the longtime leader in cryonics, Alcor was still a small nonprofit. It had been freezing the bodies and brains of its members, with the idea of one day bringing them back to life, since 1976.

The foundation, and cryonics in general, had long survived outside of mainstream acceptance. Typically shunned by the scientific community, cryonics is best known for its appearance in sci-fi films like 2001: A Space Odyssey. But its adherents have held on to a dream that at some point in the future, advances in medicine will allow for resuscitation and additional years on Earth. Over decades, small, tantalizing developments in related technology, as well as high-profile frozen test subjects like Ted Williams, have kept the hope alive. Today, nearly 200 dead patients are frozen in Alcors cryogenic chambers at temperatures of 196 C, including a handful of celebrities, who have paid tens of thousands of dollars for the goal of possible revival and ultimately reintegration into society.

But its the recent involvement of Yinfeng that signals something of a new era for cryonics. With impressive financial resources, government support, and scientific staff, its one of a handful of new labs focused on expanding the consumer appeal of cryonics and trying anew to bring credibility to the long-disputed theory of human reanimation. Just a year after Drake came on board as research director of the Shandong Yinfeng Life Science Research Institute, the subsidiary of the Yinfeng Biological Group overseeing the cryonics program, the institute performed its first cryopreservation. Its storage vats now hold about a dozen clients who are paying upwards of $200,000 to preserve the whole body.

Still, the field remains rooted in faith rather than any real evidence that it works. Its a hopeless aspiration that reveals an appalling ignorance of biology, says Clive Coen, a neuroscientist and professor at Kings College London.

Even if one day you could perfectly thaw a frozen human body, you would still just have a warm dead body on your hands.

The cryonics process typically goes something like this: Upon a persons death, a response team begins the process of cooling the corpse to a low temperature and performs cardiopulmonary support to sustain blood flow to the brain and organs. Then the body is moved to a cryonics facility, where an organ preservation solution is pumped through the veins before the body is submerged in liquid nitrogen. This process should commence within one hour of deaththe longer the wait, the greater the damage to the bodys cells. Then, once the frozen cadaver is ensconced in the cryogenic chamber, the hope of the dead begins.

Since its beginnings in the late 1960s, the field has attracted opprobrium from the scientific community, particularly its more respectable cousin cryobiologythe study of how freezing and low temperatures affect living organisms and biological materials. The Society for Cryobiology even banned its members from involvement in cryonics in the 1980s, with a former society president lambasting the field as closer to fraud than either faith or science.

In recent years, though, it has grabbed the attention of the libertarian techno-optimist crowd, mostly tech moguls dreaming of their own immortality. And a number of new startups are expanding the playing field. Tomorrow Biostasis in Berlin became the first cryonics company in Western Europe in 2019, for example, and in early 2022, Southern Cryonics opened a facility in Australia.

More researchers are open to longer-term, futuristic topics than there might have been 20 years ago or so, says Tomorrow Biostasis founder Emil Kendziorra.

Here is the original post:
Why the sci-fi dream of cryonics never died - MIT Technology Review

What Is CRISPR Gene Editing and How Does It Work?

In 2013, two biochemists published a paper proclaiming theyd discovered a potentially game-changing method of manipulating genes. CRISPR which sounds like a veggie-forward gastro pub won them each a Nobel Prize.

In the years since, CRISPR (or Clustered Regularly Interspaced Short Palindromic Repeats) has lived up to the hype. Its altered the global scientific landscape and raised questions about what kinds of revolutionary changes scientists and healthcare providers could and should pursue.

What if we could make foods allergy-free and crops drought-resistant? What if we could eliminate invasive species and protect against infectious diseases like malaria? What if we could revive extinct species? What if we could remove or repair mutations that cause inherited conditions? Or create custom immunotherapies to treat an individuals cancer?

The prospects are that exciting.

If your understanding of genetics starts and ends with high school biology or the (very fictional) Jurassic Park movies youre not alone. This stuff is complicated. Thats why we asked genomics and immunotherapy expert Timothy Chan, MD, PhD, to break CRISPR down for us, so we can better understand why, over a decade later, its still got researchers so excited.

Before we jump into CRISPR, lets start with the concept of gene editing.

Gene editing is the process of altering genetic material (DNA). That could mean changing a few individual genes or an entire sequence. Research has been ongoing for more than a decade thats looking at using gene editing on mutations that cause serious health conditions in people. The goal of this gene editing research is to eliminate or correct the mutation thats causing the health condition, or has the potential to cause one, such as certain cancers. In other research studies, gene editing is being explored so a mutation isnt passed down to children at birth.

For example, the U.S. Food and Drug Administration (FDA) approved a gene therapy in late 2022 that introduces a gene needed for blood clotting into people with hemophilia B. Its one of several cellular and gene therapy products currently in use today.

There are many different techniques and applications for gene editing. CRISPR is one approach to gene editing thats showing promise in ongoing clinical trials.

Now that were clear on what gene editing is, lets focus on a specific approach: CRISPR.

Clustered Regularly Interspaced Short Palindromic Repeats, otherwise known as CRISPR, was originally identified in bacteria, as a bacterial defense system, says Dr. Chan.

Thats right. Bacteria have immune systems, too.

CRISPR contains spacers sequences of DNA left over from unfriendly viruses or other entities as well as repeating sections of genetic material. Those sequences provide acquired immunity, and form the building blocks of the gene editing system or process. It creates a sort of blueprint that allows enzymes in genetic material to make changes to sequences of DNA in living cells. One of the best-known enzymes used for this purpose is called Cas9, which is why youll sometimes hear people talk about CRISPR-Cas9.

Over the years, people have discovered that specific enzymes that allow CRISPR to work Cas9 is one of them.But there are other ones, and they can be tailored to target sequences of interest in the DNA for specific cuts to be made, Dr. Chan explains.

You can think of the underlying mechanism of CRISPR gene editing as being similar to the way magnetic shapes are drawn to each other or the way Lego blocks fit together.

The segments in CRISPR are transcribed into RNA. This RNA includes a guide sequence, which is a match to existing DNA in a persons body.

That guide sequence can be tailored to whatever you want, Dr. Chan says. And as a result, you can make specific alterations or mutations in a part of the genome that you are targeting with a high degree of accuracy.

Along for the ride with this guide sequence is an enzyme like Cas9.

When the guide sequence and enzyme find the desired DNA to edit, the enzyme can then get down to business. It attaches itself to this DNA and makes changes, whether thats a cut or alteration.

CRISPR technology has come a long way, Dr. Chan says. The first generation of CRISPR was a great way to inactivate genes. It only made a break in genes. Then, the DNA would get filled up with natural repair enzymes.

But new versions of CRISPR like CRISPR prime or CRISPR HD are more advanced.

These can allow actual replacements to occur, Dr. Chan continues. You can even very accurately replace one sequence one of the letters in the genome with another letter. And you can make specific mutations.

CRISPRs ability to make very specific, very small cuts has the potential to transform how healthcare providers can address certain genetic diseases.

Dr. Chan is optimistic about the future of CRISPR based on the success of ongoing clinical trials in human subjects. For any type of genetic diseases caused by a single mutated gene, you can use CRISPR to mutate it and make it normal. Thats why its useful. Its a way for us to change errors in the genome.

Right now, CRISPR is geared toward correcting a single change in genes, he adds. While combinations may be possible in the future, were just not there yet.

While gene editing is already in use, CRISPR is still in the clinical trials phase, Dr. Chan says. Its used all the time in research laboratories and industries, he notes. Many clinical trials are testing CRISPR in the setting of genetic diseases and cancer.

Interestingly, CRISPR can be used to detect certain diseases. The best-known example is the Sherlock CRISPR SARS-CoV-2 Kit: A COVID-19 test that received emergency use authorization (EAU) from the FDA in 2020.

But theres no FDA-approved CRISPR therapy right now. The clinical trials are ongoing, he says.

These include trials looking at CRISPR to correct genetic diseases such as cystic fibrosis, Huntingtons disease and muscular dystrophy.

Dr. Chan adds that there are also major clinical trials in process for blood disorders, where CRISPR is being used to correct the gene alteration that causes the condition. As one example, he cites a promising trial looking at CRISPR-Cas9 gene editing for sickle cell disease and -thalassemia, written about in an early 2021 issue of the New England Journal of Medicine. -thalassemia is an inherited blood disorder that impacts the bodys ability to create hemoglobin an iron-dense protein that serves as the primary ingredient in red blood cells.

There are also clinical trials looking to see if CRISPR can be used to treat certain cancers. Dr. Chan notes that chimeric antigen receptor (CAR)T-cell therapy is one of the first gene therapies approved for leukemias. Current research is looking at whether CRISPR technology can make this treatment even more effective.

In CAR T-cell therapy, you take out T-cells from someone and put in a receptor a new way for these cells to target something on cancer cells and then put these cells back in the patient, he explains. Researchers are running trials now where they use CRISPR to alter those T-cells to make them even more active.

CRISPR therapies can take on many different forms. CRISPR has been inserted directly into the body before. It was famously injected into the eyes of seven people with a rare hereditary blindness disorder in 2020, two of whom later told NPR that they regained some ability to see colors. There are human trials in process right now that deliver CRISPR through gels and creams, through food or drink, skin grafts or injections. Ex-vivo delivery is also common: Thats when CRISPR is used to modify a cell outside the body. The cells are then re-inserted into the body using a harmless virus.

The results have been promising so far. I do believe in the next three to five years possibly even sooner were going to see approval to treat some diseases, Dr. Chan states.

With any type of CRISPR therapy, Dr. Chan says theres a risk of getting off-target effects or unexpected side effects.

Whenever youre altering something as fundamental as DNA, you just dont know what might happen he explains. Theres always a chance for the unexpected. You can potentially have effects on your DNA that were not intended.

At the moment, he doesnt have any specific examples of what these effects might be and he notes that existing research suggests the risk is pretty low. Still, data from future research might tell a different story.

Dr. Chan nevertheless sees a lot of potential for CRISPR in the coming years.

The field is moving very quickly, he says. Were seeing continual improvement of the actual CRISPR tools being used.

Its getting more accurate and more flexible in terms of what you can do. There are various engineered modified variants of CRISPR now that are allowing very specific, very accurate changes with fewer off-target effects. So, I think the future is very bright.

View original post here:
What Is CRISPR Gene Editing and How Does It Work?

Archives