Saying farewell to an old friend

There are some people who, when you think of them, always bring a smile to your face. Dr. Bert Lubin was one of those people. Sadly, we lost Bert to brain cancer two days ago. But the impact he had, not just as an advocate for stem cell research but as a pioneer in sickle cell disease research and a champion for children’s health, will live on.

Bert had a number of official titles but probably the one he was most proud of was President & CEO of Children’s Hospital Oakland (now UCSF Benioff Children’s Hospital Oakland). But it wasn’t the title that he cared about, it was the opportunity it gave him to make a difference in the life of children in Oakland, to create a program to find new treatments and cures for a life-threatening disease. And he has made a difference.

As I started to write this tribute to Bert, I thought about who I should ask for a quote. And then I realized I had the perfect person. Bert himself. I was fortunate enough to interview him in December 2018, when he decided to step down after eight years on the CIRM Board.  As always, he had his own positive spin on that, saying: “I don’t see myself leaving. I’m just repurposing what is my role in CIRM. I’m recycling and reinventing.”

And Bert was always full of invention.

He grew up in Bellevue, a small town outside Pittsburgh, PA. His parents ran a fruit and vegetable market there and, growing up, Bert often worked in the store. It wasn’t something he enjoyed but he said he learned some valuable lessons.

“I think what happened in my childhood is that I learned how to sell. I am a salesman. I hated working in that store, I hated it, but I liked the communication with people, they trusted me, I could sell things and they were good things. Like Christmas. I’m Jewish, we were the only Jews in that community, and at Christmas we sold Christmas trees, but the trees were sometimes crooked and they were $2.99 a tree so I convinced families that I could go to their house and set the tree so it looked straight and I helped them decorate it and they loved it.”

He said, thinking back on his life it’s almost as if there were a plan, even if he wasn’t aware of it.

“I started thinking about that more recently, I started wondering how did this even happen? I’m not a religious person but it’s almost like there’s some fate. How did I get there? It’s not that I planned it that way and it’s certainly not that my parents planned it because I was the first in my family to go to high school let alone college. My parents, when I went to medical school and then decided I wanted to spend more time in an academic direction, they were upset. They wanted me to go into practice in a community that I grew up in and be economically secure and not be on the fringe in what an academic life is like.”

And then, fate stepped in and brought him to the San Francisco Bay Area.

“What happened was, I was at the University of Pennsylvania having trained at Boston Children’s and Philadelphia Children’s, where I had started a sickle cell disease program, and was asked to look at a job in southern California to start a sickle cell program there. So, I flew to San Francisco because a lot of people I’d studied with were now working at UCSF and I thought it would be fun to see them before going down to southern California. They took me out to dinner and showed me around and I said this place is beautiful, I can play tennis out here all year round, there’s lots of music – I love jazz – and they said ‘you know Bert, have you looked at Oakland Children’s hospital? We want to start a sickle cell program center, but the patients are all in Oakland and the patient population that would be served is in Oakland. But if you came out to the Bay Area we could partner with you to start that program. 

“So, when I walked in the door here (at Oakland) and said ‘I want to create this northern California sickle cell center with UC’ the staff that was here said ‘you know we’re not a research hospital, we are a community based hospital’. I said, ‘I’m not saying you shouldn’t be that but I’m trying to create an opportunity here’ and they said to me ‘as long as you don’t ask for any money you can go and do whatever you want’.

‘They recognized that I had this fire in me to really create something that was novel. And the warmth and community commitment from this place is something that attracted me and then allowed me to build on that.

“For example, when I became the director of the research program we had $500,000 in NIH grants and when I left we had $60 million. We just grew. Why did we grow? Because we cared about the faculty and the community. We had a lovely facility, which was actually the home of the Black Panther party. It was the Black Panthers who started screening for sickle cell on street corners here in Oakland, and they were the start of the national sickle cell act so there’s a history here and I like that history.

“Then I got a sense of the opportunities that stem cell therapies would have for a variety of things, certainly including sickle cell disease, and I thought if there’s a chance to be on the CIRM Board, as an advocate for that sickle cell community, I think I’d be a good spokesperson. So, I applied. I just thought this was an exciting opportunity.

“I thought it was a natural fit for me to add some value, I only want to be on something where I think I add value.”

Bert added value to everything he did. And everyone he met felt valued by him. He was a mentor to so many people, young physicians and nurses, students starting out on their careers. And he was a friend to those in need.

He was an extraordinary man and we are grateful that we were able to call him a colleague, and a friend, for as long as we did.

When Burt stepped down from Children’s his colleagues put together this video about his life and times. It seems appropriate to share it again and remind ourselves of the gift that he was to everyone fortunate enough to know him.

You never forget your first

Jake Javier: Photo Michael Clemens “Sees the Day” Photography

Now that we have 64 clinical trials that we have funded (plus a few dozen more where we supported the early stage research) it’s sometimes hard to remember the details of each trial. But there is one you never forget. The very first clinical trial you funded. And we just got some encouraging news about it.

Way back in 2011 CIRM funded a clinical trial with a company called Geron, targeting spinal cord injuries. It was not only the first clinical trial we funded, it was also the first clinical trial involving the use of embryonic stem cells that was approved by the US Food and Drug Administration (FDA).

But in November of that year Geron decided to change its business plan and canceled the trial. We got all our money back – plus interest – but it was still terribly disappointing to us and to everyone who had hopes the research would help people with severe spinal cord injuries.

Fast forward three years and a company called Asterias picked up where Geron left off, getting permission from the FDA to run a clinical trial using  the same approach for spinal cord injuries. Once again CIRM funded the project.

The results from this Phase 1 trial were, to say the least, encouraging

We profiled two of the patients treated in this group who seemed to benefit a lot from the therapy; Jake Javier and Kris Boesen.

Kris Boesen

But after the initial trial it felt like someone hit the pause button. Asterias was bought up by BioTime which changed its name to Lineage Cell Therapeutics and moved much of the OPC1 spinal cord injury program to Israel. Then last week Lineage announced it was unpausing the program.

In a news release they announced that by moving the program to their cGMP manufacturing plant in Israel they were able to make “process improvements” in the program and, more importantly; “ Lineage intends to meet with the U.S. Food and Drug Administration (FDA) to discuss further development of the OPC1 program by the end of 2020.”

Brian M. Culley, Lineage’s CEO said:  “We have worked diligently over the past year to transition all manufacturing activities for the OPC1 program to our in-house cGMP facility, where our experienced cell therapy production team could develop and deploy much-needed improvements and modernization to the production and analytical processes. This work has achieved significantly better efficiency and improved quality control, which we expect will enable a consistent supply of material to support a late-stage clinical trial of OPC1. With these necessary steps now completed, our focus turns to developing a “thaw-and-inject” formulation and superior delivery tools, to enable an easier surgical procedure and facilitate faster enrollment in the next clinical trial. We also are evaluating ways to return OPC1 to the clinic sooner than originally planned, reflecting our view of compelling clinical data which continues to read out from the 25-patient phase 1/2a SCiStar study.”

So, almost a decade after we first became involved with this project, we’re happy to say it’s alive and seemingly well and getting ready to take the next step in helping people with spinal cord injuries. We’ll let you know how it goes.

One last thing. One of the reasons we are such fans of the approach is Jake Javier. We have come to know and admire him and watch him fight back from his injury. He is a remarkable young man in many ways. He is now a student at Cal Poly where they made this video about him.

CIRM Board Approves Third Clinical Trial for COVID-19

Dr. Xiaokui Zhang (left), Dr. Albert Wong (center), and Dr. Preet Chaudhary (right)

Today the governing Board of the California Institute for Regenerative Medicine (CIRM) awarded $750,000 to Dr. Xiaokui Zhang at Celularity to conduct a clinical trial for the treatment of COVID-19.  This brings the total number of CIRM clinical trials to 64, including three targeting the coronavirus.

This trial will use blood stem cells obtained from the placenta to generate natural killer (NK) cells, a type of white blood cell that is a vital part of the immune system, and administer them to patients with COVID-19.  NK cells play an important role in defense against cancer and in fighting off viral infections.  The goal is to administer these cells to locate the active sites of COVID-19 infection and destroy the virus-infected cells.  These NK cells have been used in two other clinical trials for acute myeloid leukemia and multiple myeloma.

The Board also approved two additional awards for Discovery Stage Research (DISC2), which promote promising new technologies that could be translated to enable broad use and improve patient care.

One award for $100,000 was given to Dr. Albert Wong at Stanford.  Dr. Wong has recently received an award from CIRM to develop a vaccine that produces a CD8+ T cell response to boost the body’s immune response to remove COVID-19 infected cells.  The current award will enable him to expand on the initial approach to increase its potential to impact the Latinx and African American populations, two ethnicities that are disproportionately impacted by the virus in California.

The other award was for $249,996 and was given to Dr. Preet Chaudhary at the University of Southern California.  Dr. Chaudary will use induced pluripotent stem cells (iPSCs) to generate natural killer cells (NK). These NK cells will express a chimeric antigen receptor (CAR), a synthetic receptor that will directly target the immune cells to kill cells infected with the virus.  The ultimate goal is for these iPSC-NK-CAR cells to be used as a treatment for COVID-19. 

“These programs address the role of the body’s immune T and NK cells in combatting viral infection and CIRM is fortunate enough to be able to assist these investigators in applying experience and knowledge gained elsewhere to find targeted treatments for COVID-19” says Dr. Maria T. Millan, the President & CEO of CIRM. “This type of critical thinking reflects the resourcefulness of researchers when evaluating their scientific tool kits.  Projects like these align with CIRM’s track record of supporting research at different stages and for different diseases than the original target.”

The CIRM Board voted to endorse a new initiative to refund the agency and provide it with $5.5 billion to continue its work. The ‘California Stem Cell Research, Treatments and Cures Initiative of 2020 will appear on the November ballot. 

The Board also approved a resolution honoring Ken Burtis, PhD., for his long service on the Board. Dr. Burtis was honored for his almost four decades of service at UC Davis as a student, professor and administrator and for his 11 years on the CIRM Board as both a member and alternate member. In the resolution marking his retirement the Board praised him, saying “his experience, commitment, knowledge, and leadership, contributed greatly to the momentum of discovery and the future therapies which will be the ultimate outcome of the dedicated work of the researchers receiving CIRM funding.”

Jonathan Thomas, the Chair of the Board, said “Ken has been invaluable and I’ve always found him to have tremendous insight. He has served as a great source of advice and inspiration to me and to the ICOC in dealing with all the topics we have had to face.” 

Lauren Miller Rogen thanked Dr. Burtis, saying “I sat next to you at my first meeting and was feeling so extraordinarily overwhelmed and you went out of your way to explain all these big science words to me. You were always a source of help and support, and you explained things to me in a way that I always appreciated with my normal brain.”

Dr. Burtis said it has been a real honor and privilege to be on the Board. “I’ve been amazed and astounded at the passion and dedication that the Board and CIRM staff have brought to this work. Every meeting over the years there has been a moment of drama and then resolution and this Board always manages to reach agreement and serve the people of California.”

A ready-made approach to tackling COVID-19

Coronavirus particles, illustration.

In late March the CIRM Board approved $5 million in emergency funding for COVID-19 research. The idea was to support great ideas from California’s researchers, some of which had already been tested for different conditions, and see if they could help in finding treatments or a vaccine for the coronavirus.

Less than a month later we were funding a clinical trial and two other projects, one that targeted a special kind of immune system cell that has the potential to fight the virus.

Our friends at UCLA have just written a terrific piece on this project and the team that came up with the idea. Here is that article.

Researchers use stem cells to model the immune response to COVID-19

By Tiare Dunlap

Cities across the United States are opening back up, but we’re still a long way from making the COVID-19 pandemic history. To truly accomplish that, we need to have a vaccine that can stop the spread of infection.

But to develop an effective vaccine, we need to understand how the immune system responds to SARS-CoV-2, the virus that causes COVID-19.

Vaccines work by imitating infection. They expose a person’s immune system to a weakened version or component of the virus they are intended to protect against. This essentially prepares the immune system to fight the virus ahead of time, so that if a person is exposed to the real virus, their immune system can quickly recognize the enemy and fight the infection. Vaccines need to contain the right parts of the virus to provoke a strong immune response and create long-term protection.

Most of the vaccines in development for SARS CoV-2 are using part of the virus to provoke the immune system to produce proteins called antibodies that neutralize the virus. Another way a vaccine could create protection against the virus is by activating the T cells of the immune system.

T cells specifically “recognize” virus-infected cells, and these kinds of responses may be especially important for providing long-term protection against the virus. One challenge for researchers is that they have only had a few months to study how the immune system protects against SARS CoV-2, and in particular, which parts of the virus provoke the best T-cell responses.

This is where immunotherapy researchers and UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research members Dr. Gay Crooks and Dr. Christopher Seet come in.

Dr. Gay Crooks: Photo courtesy UCLA

For years, they have been perfecting an innovative technology that uses blood-forming stem cells — which can give rise to all types of blood and immune cells — to produce a rare and powerful subset of immune cells called type 1 dendritic cells. Type 1 dendritic cells play an essential role in the immune response by devouring foreign proteins, termed antigens, from virus-infected cells and then chopping them into fragments. Dendritic cells then use these protein fragments to trigger T cells to mount an immune response.

Dr. Christopher Seet: Photo courtesy UCLA

Using this technology, Crooks and Seet are working to pinpoint which specific parts of the SARS-CoV-2 virus provoke the strongest T-cell responses.

Building long-lasting immunity

“We know from a lot of research into other viral infections and also in cancer immunotherapy, that T-cell responses are really important for long-lasting immunity,” said Seet, an assistant professor of hematology-oncology at the David Geffen School of Medicine at UCLA. “And so this approach will allow us to better characterize the T-cell response to SARS-CoV-2 and focus vaccine and therapeutic development on those parts of the virus that induce strong T-cell immunity.”

This project was recently awarded $150,000 from the California Institute for Regenerative Medicine, the state’s stem cell agency. The award was matched by the UCLA Broad Stem Cell Research Center.

Crooks’ and Seet’s project uses blood-forming stem cells taken from healthy donors and infected with a virus containing antigens from SARS-CoV-2. They then direct these stem cells to produce large numbers of type 1 dendritic cells using a new method developed by Seet and Suwen Li, a graduate student in Crooks’ lab. Both Seet and Li are graduates of the UCLA Broad Stem Cell Research Center’s training program.

“The dendritic cells we are able to make using this process are really good at chopping up viral antigens and eliciting strong immune responses from T cells,” said Crooks, a professor of pathology and laboratory medicine and of pediatrics at the medical school and co-director of the UCLA Broad Stem Cell Research Center.

When type 1 dendritic cells chop up viral antigens into fragments, they present these fragments on their cell surfaces to T cells. Our bodies produce millions and millions of T cells each day, each with its own unique antigen receptor, however only a few will have a receptor capable of recognizing a specific antigen from a virus.

When a T cell with the right receptor recognizes a viral antigen on a dendritic cell as foreign and dangerous, it sets off a chain of events that activates multiple parts of the immune system to attack cells infected with the virus. This includes clonal expansion, the process by which each responding T cell produces a large number of identical cells, called clones, which are all capable of recognizing the antigen.

“Most of those T cells will go off and fight the infection by killing cells infected with the virus,” said Seet, who, like Crooks, is also a member of the UCLA Jonsson Comprehensive Cancer Center. “However, a small subset of those cells become memory T cells — long-lived T cells that remain in the body for years and protect from future infection by rapidly generating a robust T-cell response if the virus returns. It’s immune memory.”

Producing extremely rare immune cells

This process has historically been particularly challenging to model in the lab, because type 1 dendritic cells are extremely rare — they make up less than 0.1% of cells found in the blood. Now, with this new stem cell technology, Crooks and Seet can produce large numbers of these dendritic cells from blood stem cells donated by healthy people, introduce them to parts of the virus, then see how T cells  taken from the blood can respond in the lab. This process can be repeated over and over using cells taken from a wide range of healthy people.

“The benefit is we can do this very quickly without the need for an actual vaccine trial, so we can very rapidly figure out in the lab which parts of the virus induce the best T-cell responses across many individuals,” Seet said.

The resulting data could be used to inform the development of new vaccines for COVID-19 that improve T-cell responses. And the data about which viral antigens are most important to the T cells could also be used to monitor the effectiveness of existing vaccine candidates, and an individual’s immune status to the virus.

“There are dozens of vaccine candidates in development right now, with three or four of them already in clinical trials,” Seet said. “We all hope one or more will be effective at producing immediate and long-lasting immunity. But as there is so much we don’t know about this new virus, we’re still going to need to really dig in to understand how our immune systems can best protect us from infection.”

Supporting basic research into our body’s own processes that can inform new strategies to fight disease is central to the mission of the Broad Stem Cell Research Center.

“When we started developing this project some years ago, we had no idea it would be so useful for studying a viral infection, any viral infection,” Crooks said. “And it was only because we already had these tools in place that we could spring into action so fast.”

Scientists Engineer Stem Cells to Fight HIV

Image of the virus that causes AIDS – courtesy NIH

If that headline seems familiar it should. It came from an article in MIT Technology Review back in 2009. There have been many other headlines since then, all on the same subject, and yet here we are, in 2020, and still no cure for HIV/AIDS. So what’s the problem, what’s holding us back?

First, the virus is incredibly tough and wily. It is constantly mutating so trying to target it is like playing a game of ‘whack a mole’. Secondly not only can the virus evade our immune system, it actually hijacks it and uses it to help spread itself throughout the body. Even new generations of anti-HIV medications, which are effective at controlling the virus, can’t eradicate it. But now researchers are using new tools to try and overcome those obstacles and tame the virus once and for all.

Dr. Scott Kitchen: Photo David Geffen School of Medicine, UCLA

UCLA researchers Scott Kitchen and Irvin Chen have been awarded $13.65 million by the National Institutes of Health (NIH) to see if they can use the patient’s own immune system to fight back against HIV.

Dr. Irvin Chen: Photo UCLA

Dr. Kitchen and Dr. Chen take the patient’s own blood-forming stem cells and then, in the lab, they genetically engineer them to carry proteins called chimeric antigen receptors or CARs. Once these blood cells are transplanted back into the body, they combine with the patient’s own immune system T cells (CAR T). These T cells now have a newly enhanced ability to target and destroy HIV.

That’s the theory anyway. Lots of research in the lab shows it can work. For example, the UCLA team recently showed that these engineered CAR T cells not only destroyed HIV-infected cells but also lived for more than two years. Now the team at UCLA want to take the lessons learned in the lab and apply them to people.

In a news release Dr. Kitchen says the NIH grant will give them a terrific opportunity to do that: “The overarching goal of our proposed studies is to identify a new gene therapy strategy to safely and effectively modify a patient’s own stem cells to resist HIV infection and simultaneously enhance their ability to recognize and destroy infected cells in the body in hopes of curing HIV infection. It is a huge boost to our efforts at UCLA and elsewhere to find a creative strategy to defeat HIV.”

By the way, CIRM helped get this work off the ground with an early-stage grant. That enabled Dr. Kitchen and his team to get the data they needed to be able to apply to the NIH for this funding. It’s a great example of how we can kick-start projects that no one else is funding. You can read a blog about that early stage research here.

CIRM has already funded three clinical trials targeting HIV/AIDS. Two of these are still active; Dr. Mehrdad Abedi at UC Davis and Dr. John Zaia at City of Hope.

Stem cells used to promote quick and precise bone healing

A close-up view of the intricate microarchitecture of the pluripotent stem-cell-derived extracellular matrix. Image Credit: Carl Gregory/Texas A&M

Although some broken bones can be mended with the help of a cast, others require more complex treatments. Bone grafts, which can come from the patient’s own body or a donor, are used to transplant bone tissue to the injury site. However, these procedures can have setbacks such as increased recovery time and chronic pain. Each year approximately 600,000 people in the United States alone experience complications from bone healing.

Researchers at Texas A&M University found a way to use induced pluripotent stem cells (iPSCs), a type of stem cell that can turn into any cell type and can be derived from adults cells (e.g. skin cells), to create superior bone grafts. The team of researchers said these grafts could potentially be used to promote swift and precise bone healing, enabling patients to optimally benefit from surgical intervention.

The Texas A&M team used iPSCS to make mesenchymal stem cells (MSCs), which make the extracellular matrix needed for bone grafts. MSCs can be obtained from bone marrow, but they have a relatively shorter life span and are not as biologically active when compared to MSCs generated from iPSCs.

To test the effectiveness of their iPSC generated bone grafts, they implanted the extracellular matrix at a site of bone defects. After a few weeks, they found that their iPSC generated matrix was five to sixfold more effective than the best FDA-approved graft stimulator.

In a news release from Texas A&M, Dr. Roland Kaunas discusses the potential benefits of using iPSC generated bone grafts.

“Our material is very promising because the pluripotent stem cells can ideally generate many batches of the extracellular matrix from just a single donor which will greatly simplify the large-scale manufacturing of these bone grafts.”

Additionally, the Texas A&M team said this approach has the potential to be incorporated into numerous engineered implants, such as 3D-printed implants or metal screws, so that these parts integrate better with the surrounding bone.

The full results of this study were published in Nature Communications.

A brief video on bone grafts from Texas A&M is available below.

Magnetized stem cells used to treat lung disease in mice

Magnetic targeting technique has emerged as a new strategy to aid delivery, increase retention, and enhance the effects of mesenchymal stromal cells (MSCs) but, so far, has not been performed in lung diseases. With the aid of magnets, magnetized MSCs remained longer in the lungs, and this was associated with increased beneficial effects for the treatment of silicosis in mice. Image Credit: AlphaMed Press

Certain jobs, such as construction work and sand blasting, are quite labor intensive but can also lead to some unexpected health complications down the road. One of these is called silicosis, a serious lung disease that affects millions of workers worldwide. It is the result of years of breathing in silica, a type of dust particle most commonly found in sand. The particles can cause inflammation and scarring of the lung tissue, which can lead to trouble breathing and death in the most severe cases. There is currently no cure for this condition and once the damage is done it cannot be reversed.

However, Dr. Patricia Rocco and Dr. Fernanda Cruz from the Laboratory of Pulmonary Investigation at Universidade Federal do Rio de Janeiro, Brazil have found a promising approach to treat silicosis that involves the use of stem cells and magnetization.

In this study, mesenchymal stromal cells (MSCs), a type of stem cell that has anti-inflammatory properties, were magnetized using specialized nanoparticles. The effects of the newly magnetized MSCs were then studied in mice in which silicosis was induced to see if magnetization could aid in delivery to the lungs. One group of mice was injected with saline (as a control study) while another group was injected with the magnetized MSCs. A third group of mice was injected with magnetized MSCs with a pair of magnets attached to their chest for 2 days. The results showed that using the magnetized MSCs alongside the magnets proved to be most effective in migrating the cells towards the lungs.

In a news release, Dr. Cruz elaborated on their findings for this portion of the study.

“Upon removal of the magnets, we examined all the animals in all the groups and found that those implanted with magnets had a significantly larger amount of magnetized MSCs in their lungs.”

For the next portion of the study, the team compared treatments in mice using magnetized MSCs with magnets vs non-magnetized MSCs. After 7 days, the magnets were removed from the mice with magnetized MSCs and their lungs were evaluated. It was found that those treated with magnetized MSCs and magnets showed significant signs of lung improvement while the other mice did not.

In the same news release, Dr. Rocco discusses the implications that these results have in terms of developing a potential treatment.

“This tells us that magnetic targeting may be a promising strategy for enhancing the beneficial effects of MSC-based cell therapies for silicosis and other chronic lung diseases.”

The full results of this study were published in Stem Cells Translational Medicine (SCTM).

CIRM has recently funded a clinical trial that uses MSCs to treat patients with acute respiratory distress syndrome (ARDS), a life-threatening lung injury that occurs when fluid leaks into the lungs, in both COVID-19 positive and COVID-19 negative patients.

Super charging killer cells to fight leukemia

Colorized scanning electron micrograph of a natural killer cell.
Photo credit: National Institute of Allergy and Infectious Diseases

Racing car drivers are forever tinkering with their cars, trying to streamline them and soup up their engines because while fast is good, faster is better. Researchers do the same things with potential anti-cancer therapies, tinkering with them to make them safer and more readily available to patients while also boosting their ability to fight cancer.

That’s what researchers at the University of California San Diego (UCSD), in a CIRM-funded study, have done. They’ve taken immune system cells – with the already impressive name of ‘natural killer’ (NK) cells – and made them even deadlier to cancers.

These natural killer (NK) cells are considered one of our immune system’s frontline weapons against outside threats to our health, things like viruses and cancer. But sometimes the cancers manage to evade the NKs and spread throughout the body or, in the case of leukemia, throughout the blood.

Lots of researchers are looking at ways of taking a patient’s own NK cells and, in the lab boosting their ability to fight these cancers. However, using a patient’s own cells is both time consuming and very, very expensive.

Dan Kaufman MD

Dr. Dan Kaufman and his team at UCSD decided it would be better to try and develop an off-the-shelf approach, a therapy that could be mass produced from a single batch of NK cells and made available to anyone in need.

Using the iPSC method (which turns tissues like skin or blood into embryonic stem cell-like cells, capable of becoming any other cell in the body) they created a line of NK cells. Then they removed a gene called CISH which slows down the activities of cytokines, acting as a kind of brake or restraint on the immune system.

In a news release, Dr. Kaufman says removing CISH had a dramatic effect, boosting the power of the NK cells.

“We found that CISH-deleted iPSC-derived NK cells were able to effectively cure mice that harbor human leukemia cells, whereas mice treated with the unmodified NK cells died from the leukemia.”

Dr. Kaufman says the next step is to try and develop this approach for testing in people, to see if it can help people whose disease is not responding to conventional therapies.

“Importantly, iPSCs provide a stable platform for gene modification and since NK cells can be used as allogeneic cells (cells that come from donors) that do not need to be matched to individual patients, we can create a line of appropriately modified iPSC-derived NK cells suitable for treating hundreds or thousands of patients as a standardized, ‘off-the-shelf’ therapy.”

The study is published in the journal Cell Stem Cell.

CIRM Board Approves Two Additional COVID-19 Projects

Dr. Vaithilingaraja Arumugaswami (left) and Dr. Song Li (right), UCLA

Today the governing Board of the California Institute for Regenerative Medicine (CIRM) approved two additional projects as part of the $5 million in emergency funding for COVID-19 related projects. This brings the number of projects CIRM is supporting to 11, including two clinical trials.

The Board awarded $349,999 to Dr. Vaithilingaraja Arumugaswami at UCLA.  The focus of this project will be to study Berzosertib, a therapy targeting viral replication and damage in lung stem cells.  The ultimate goal would be to use this agent as a therapy to prevent COVID-19 viral replication in the lungs, thereby reducing lung injury, inflammation, and subsequent lung disease caused by the virus.  

This award is part of CIRM’s Translational Stage Research Program (TRAN1), which promotes the activities necessary for advancement to clinical study of a potential therapy.

The Board also awarded $149,916 to Dr. Song Li at UCLA.  This project will focus on developing an injectable biomaterial that can induce the formation of T memory stem cells (TMSCs), an important type of stem cell that plays a critical role in generating an immune response to combat viruses. In vaccine development, there is a major challenge that the elderly may not be able to mount a strong enough immunity.  This innovative approach seeks to address this challenge by increasing TMSCs in order to boost the immune response to vaccines against COVID-19.

This award is under CIRM’s Discovery Stage Research Program (DISC2), which promotes promising new technologies that could be translated to enable broad use and improve patient care.

“CIRM continues to support novel COVID-19 projects that build on previous knowledge acquired,” says Dr. Maria T. Millan, the President & CEO of CIRM. “These two projects represent the much-needed multi-pronged approach to the COVID-19 crisis, one addressing the need for effective vaccines to prevent disease and the other to treat the severe illness resulting from infection.”

The growth of virtual clinical trials during COVID-19

A participant in a virtual study run by the California firm Science 37 receives materials at home. Credit: Christian Alexander

In the midst of the coronavirus pandemic, there has been a desire to continue to conduct ongoing clinical trials while maintaining social distancing as much as possible. Clinical trial participants have been hesitant to attend routine check-ups and monitoring due to the risk of exposure and health-care workers are stretched beyond their capacity treating COVID-19 patients. As a result of this, many clinical trials have been put on hold.

Since the coronavirus began to spread, Science 37, a company that supports virtual clinical trials conducted mostly online, began to receive hundreds of inquiries every week from pharmaceutical companies, medical centers, and individual investigators. These inquiries revolve around how best to transition to a virtual clinical trial structure, where consultations are performed online and paperwork and data are collected remotely as much as possible.

In an article published in the journal Nature, Jonathan Cotliar, chief medical officer of Science 37, discusses the impact that COVID-19 has had on the company.

“It’s exponentially accelerated the adoption curve of what we were already doing. That’s been a bit surreal.”

One example of a virtual clinical trial was conducted at the University of Minnesota in Minneapolis by Dr. David Boulware and his colleagues. They conducted a randomized, controlled, virtual trial of the malaria drug hydroxychloroquine to find out if it was effective at protecting people from COVID-19 (the results found that it was not). The trial included more than 800 participants and sent them medicine by FedEx delivery while monitoring their health via virtual appointments.

It is anticipated that even as the coronavirus pandemic and social distancing measures come to an end, virtual clinical trials will continue to be used in the future. Patient advocates have long pushed for these kinds of trials to ease the burden of clinical trial participation, which tends to be more challenging for underrepresented and underserved communities. As a result of the increase in virtual trials, the FDA has released guidelines for conducting virtual trials in order to streamline the process. It is possible that virtual trials might speed up enrollment of participants, which could help speed up the drug-development process while still maintaining rigorous standards.