The future of cancer treatments is bright with the promise of personalized care. In this conversation, Dr. Laura Towart shares some of the things they have been developing in the world of cancer treatments. She talks about the challenges small startups face when attempting to make significant breakthroughs in the field of personalized treatments, highlighting the need for more substantial funding and collaborative efforts among companies. She also emphasizes the willingness of many patients, especially those with advanced or hard-to-treat cancers, to volunteer for experimental treatments and therapies, driven by the hope of advancing medical knowledge and helping future patients. Additionally, she touches on the rapid advancement of genetic sequencing technology, its role in understanding individual diseases, and the potential of genetics in revolutionizing healthcare in the near future. This conversation demonstrates the growing importance of personalized medicine and the potential for genetic insights to transform healthcare approaches. Tune in for more!

Watch the episode here

 

Listen to the podcast here

 

The Future Of Cancer Treatments – A Conversation With Dr. Laura Towart

In this episode, we have Laura Towart with us, who is the CEO and Founder of Vivan Therapeutics who’s doing a lot of interesting stuff in the cancer and genetic space. I do want to talk with you a little bit about both of those topics but tell us a little bit about yourself, how you got into this, and what you’re doing.

Thank you so much for inviting me to be here. I am originally from New York. I’m a scientist by training. I was doing my PhD at Cornell and Sloan Kettering many years ago. I left my studies to found my first company, which is called Celmatix. It started as a fertility genomics company and has evolved into an early-stage therapeutics company for women’s health.

Over the years, I’ve left New York City and lived in different places around the world. I became a little bit separated from Celmatix because of the distance and my travels. I was introduced to this technology that underlies Vivan Therapeutics by the head of commercialization at Mount Sinai Medical Center. My main passion is personalized medicine and genomics is my specialty. When I heard about this technology, I got excited about the opportunity to personalize cancer treatment and all genetic diseases. I started working with Mount Sinai. It’s been over a decade. I launched Vivan Therapeutics here in London in 2018.

I feel like that’s an interesting thing that people are talking about, which is personalized medicine but it hasn’t trickled down to any therapeutics that I can name. Is that inaccurate? Are there any therapeutics that are out there on the market that people might have some experience with that have been shown to be effective?

Yes. For breast cancer, you have the HER2 family. You’ve got some personalization happening. There’s a lot further we can go. Having dealt with a family member with cancer, I was surprised by the lack of personalization despite the progress that we’ve made.

 

FSP-DFY 22 | Cancer Treatments

 

Cancer is such a terrible disease. Both my parents passed from cancer so I am understanding of people who are going through that struggle. It’s not a very good outcome if you have certain types of cancer.

My mother passed out of cancer. It’s been very difficult being on both sides working in this space for so long and then seeing the realities in the clinic.

What cancer did your mom have?

Colorectal cancer, which is the cancer that we focus on at Vivan.

Tell me how Vivan Therapeutics would affect colorectal cancer.

We licensed the technology in partnership with Mount Sinai so Mount Sinai owns a portion of the company. Mount Sinai pioneered this. When I became involved, the technology was already fully developed and was successful in clinical trials. I came in very late. The way that the technology works is a patient would go to their oncologist anywhere in the world. Their tumor would be biopsied as standard, a blood sample would be taken, and then we coordinate whole exome sequencing of the patient’s tumor and blood.

We have a proprietary variant calling software that reads through the exome and identifies the mutations that are causing that person’s cancer. What makes us unique is that we look at not just the main drivers but also passenger mutations and things that might help the cancer evade therapeutics so they help form resistance.

We engineer all of these mutations into fruit flies. We create a fruit fly avatar. We do this in a tissue-specific way. If the patient has colorectal cancer, we engineer the tumor to develop in the lowermost portion of the gut of the fly. We then create what we call the avatar army. We expand that fly population to any number, usually half a million for each patient. We use that army to screen all approved drugs.

We screen everything ever approved by the FDA, which is about 2,000 drugs alone and in combination until we find the right combination to rescue the fly because the fly is going to have a lethal tumor. It will die in a controlled period unless it’s rescued. We identify these never-before-seen cocktails. It’s usually a combination of cancer and non-cancer drugs, which makes it exciting. They’re small molecules so it’s usually very targeted, cheaper, and less toxic. We’re getting these cool combinations which we then also patent and looking to further develop.

Does this remove the need for surgery?

No. Surgery is a big part of it as well. Usually, it’s a combination of surgery and chemotherapy. It includes chemotherapy usually but it makes it much more personalized.

You were saying that there are applications outside of cancer.

Mount Sinai has shown two things. They’ve done a lot of work in modeling rare genetic diseases for therapeutic discovery. Those are diseases where there isn’t a standard of care or therapy. We’re modeling ECHS1, which is a rare genetic disease. We’ve created a working model and we’re working towards therapeutic discovery.

We could also do something cool. We could model diabetes and cancer in the same animal so we can model comorbidities. Mount Sinai has published that patients who have diabetes and cancer should get different treatments than patients with cancer alone. That’s something that we’re going to be working to explore clinically.

Could this work with craniofacial syndromes? Let’s say someone is born with some sort of genetic disease. My background is in craniofacial disorders. Let’s say cherubism. It is a disease that these young individuals will grow tumors in their faces that will cause them to have further complications later on in their lives. Can you model that disease and then find existing drugs that target that?

I’m not sure. If it has anything to do with bone formation, we could model it well. It has to have a homolog in the fly. ECHS1 is a metabolic disease. There is a homolog. That’s why we are able to model it. With the way that we screen the drugs, there has to be a pretty strong phenotype so that we can see a reversal of that. Usually, with cancer, we use lethality as the screen. It’s very simple. Either the fly is dead or alive. With something like that, it’s a bit more nuanced so we’d have to be able to see.

We’ve invented this new image capture and analysis software. We’re saying, “Is the fly dead or alive,” in some crude way. It’s more refined than that. What we’re able to do is measure over time and say, “The quality of the eye of the fly is a measure of health.” We can, as we’re developing the technology, further look to see we’re not just looking at survival. We’re looking at metrics of health. That’s when we can start getting into cool things like looking at longevity and quality of life.

Even college students are using fruit flies. Is that the only thing available or are there other similar animal models that people are using that are more analogous to humans?

In terms of models, there are always advantages and disadvantages. The advantage of the fruit flies is that we’re able to engineer any number of genetic alterations quickly and cheaply. For example, in a mouse model, you can maybe engineer 3 to 4 mutations. It will take a long time and then you won’t have a large number of animals to screen. You could usually say, “There are three standard-of-care therapies. Which one is best?” The mouse might be good for that but you’re not modeling the full complexity. The basic premise underlying the development of our technology is that cancer is complex. There are so many mutations driving cancer.

The promise of targeted therapy where we identify a single mutation and drug it with one drug failed. There was a great promise for that. Studies have shown it makes no difference to target one because it’s like HIV where you have multiple leading heads. If you’re going to hit it with one therapy, it’s going to mutate and evolve around it very quickly. That’s why we have patients that have an immediate response and then they have an immediate remission.

We know that you need to have this combination front but to do that, you have to have complex models to be able to understand how that’s working. Ours is very good in that it’s complex. We’re able to get complex tumors. We can screen so many combinations that you can’t do in any other system. It’s a great first step in identifying things that you wouldn’t even think about.

 

 

We don’t presume to understand and say, “I want to test this or this.” We asked the oncologist. If the oncologist wants to test a certain thing or combination, we’ll test it but we’ll show them how what they choose ranks against what the kitchen sink approach would show. We’re able to uncover these cool things that you wouldn’t find any other way. Organoids are a big deal. Everyone’s talking about them. In terms of relevance, there are a few problems with organoids. It’s difficult. Not every patient’s tissue will be cultured well to test it.

Organoids are artificial organs created by a patient. Is that it?

Yes. It could be from a patient. You could buy commercial ones. The idea is that you’re testing sometimes the tissue from an actual patient. Even though cancer grows and proliferates very well in a body, it doesn’t necessarily proliferate well in culture. It becomes a little artificial because the tumor is communicating with other organs, immune cells, and the microbiome. It’s much bigger than the way that we simplify it in culture. There are advantages. Organoids are getting better but at this point, I don’t think it’s better than our technology. It’s complimentary.

Has your technology objectively been shown to increase survival rates or have a better effect on existing models or therapies?

We don’t have a large enough dataset to be comparative but we have published case studies where we’ve shown patients who have failed every other therapy that had no other option had dramatic responses.

We have published case studies where we’ve shown patients that have failed every other therapy that had no other option had dramatic response.

These patients, were they all colorectal cancer or all sorts of other cancers?

A few of the patients were colorectal, one published, and then another patient was adenocarcinoma.

The therapy itself, I could see it being used in metabolic diseases and other genetic diseases. One of the things that you highlighted a little bit is other further applications. Tell me a little bit about what your goal is outside of cancer other than metabolic diseases. What do you think this therapy could have an effect on our health?

We’re hitting the tip of the iceberg. We can model some neurodegenerative diseases for therapeutic discovery. We’re treating one disease when a patient has multiple and we’re treating them all separately. The biggest advance in what our technology is able to offer is the ability to model more than one at once. That’s going to be the main transformation. When you could take a fecal swab, culture that to the actual microbiome of the patient, look more into the epigenetics of a patient, and create a full model of multiple things happening at once is going to be the future.

 

 

It’s such an interesting and technically complicated therapy that you’re talking about. Genetics in general is this thing that you talk about and is this buzzword that people think they understand but don’t really understand. Honestly, even as somebody in medicine, I don’t feel like I understand it the way that I should. When we talk about the human genome being sequenced and all of these breakthroughs that have happened over the years, there was always this promise like, “We’re going to figure it out. All these things are going to change.” I don’t feel like that’s trickled down.

I’m quite frustrated, to be honest. When I started Celmatix, which was in 2008, we did the first whole genome sequence of idiopathic in fertile women. We did it at Cornell, which at the time, was the number one center for IVF in the world. We identified so many biomarkers that could help guide a woman through her fertility journey. We developed a test called the Fertilome Test. It was much more advanced than anything that’s developed. We were ahead of the market because doctors weren’t using it and no one understood what to make of it. It was a pioneer. 2008 is a long time ago.

When I’m talking to oncologists and I see oncologists in major centers, the most that they’re doing for patients is standard. It’s a 70-mutation panel for cancer. There’s so much more information beyond it. It’s almost better not to do the 70-mutation. Why get a little slice of the picture? Even when they do that, a lot of the time, they’re not making any decision based on that. It’s a little information, not enough to make a decision. I feel like there’s a lot of waste.

I’m very happy to be in the UK because the UK invested a lot in Genomics England and whole genome sequencing patients with cancer. There’s a huge data set. We put a lot of money into sequencing patients and all the data is sitting there. We need to fund people that could make that information actionable. We’ve gotten a lot of funding because of that. It’s been helpful. There is a major gap in information. Even the information that we have will be drastically different. We don’t even know what to make of a lot of the non-coding regions and how it influences them.

Epigenetics, everybody’s talking about it but nobody has any idea of what to do that with. I can see the roadblocks from a provider’s perspective. As somebody who’s on the front lines, I don’t want to do anything that hasn’t been tried and tested and has been drilled into me through the literature as a standard course of treatment.

Even so, I wonder if there are some other social factors that you have insight into because there is a disconnect between research and medicine. You see things differently than I see things. The adoption of new technology is going to be difficult unless it’s very lucrative to be quite honest with you. When the new facial serum is coming out, like the vampire facial with PRP or something like that, that’s going to spread like wildfire. I wonder from your perspective what are some of the roadblocks that you see. I see myself being set in my ways as one and the financial incentives as another. You might see some other things.

It’s a very interesting topic. I’ve had discussions with people from different walks of life and different perspectives in terms of policy, economics, and medicine. The ethics of treating patients with something new is a big conversation to have. Imagine you’re a patient and you have stage four cancer. You’re going through therapies that are painful and that you’re not responding to. You almost don’t want to get treated anymore.

You then do a test like ours. You pay for it out of pocket. It costs $15,000. It includes the sequencing and the test itself. You have a personalized recommendation. It’s given you hope. Your oncologist has recommended you to try us. The oncologist brings our report to the tumor board. In some cases, the tumor board accepts it. In the UK, some medical centers have a genome board.

In our case, there’s someone on the genome board who doesn’t agree. One single human is blocking this patient’s access to trying this therapy that they could get from the pharmaceutical company via compassionate use. Pharmaceutical companies are reading our reports and buying into our rationale of, “You’ve modeled this. Our therapy is helpful. The patient isn’t responding.” It’s not an element of cost. It’s an element of access. At this point, it’s an ethical and risky question. It goes to the medical center but they can’t do it unless it passes through the board. 

This patient is a prominent media person. He was like, “I want to make this public. I want to kick up a fuss. Why is one person blocking me from being able to get a treatment that I’d sign any liability form for? I want to get this treatment.” We were able to get the patient the treatment in Switzerland, Germany, and America but he can’t get it in the UK, not at the center.

We have all of these elements of personal choice and freedom. It’s a combination of approved drugs. The drugs are approved. I don’t remember in this patient’s case. In most cases, it’s an asthma medicine, a cancer drug, or a psoriasis medicine. Some patients could be in these combinations by having two diseases. One’s an anti-hypertensive.

It is crazy that you have these people who are gatekeepers. They don’t want to tell someone who’s going through something that they can’t imagine that they can’t have something that could help them. To do that, they’d have to travel. For patients with colorectal cancer, it’s very hard to get on a plane. They’re not very mobile. It’s difficult.

It’s been difficult as the person offering the therapy because we didn’t imagine those difficulties of patients getting access. I feel bad when someone pays for something that they can’t get. I’ve given them some glimmer of hope that they can have something that can help but it’s going to be difficult to get it. We’ve been trying to take a stand on this.

We’re not well-funded but if we were, we have a documentary filmmaker that we want to have come and interview patients, oncologists, and tumor board members. We want to create this social message and bring awareness to this thing. It’s not just our technology. It’s others. Who determines who can access what?

I had this conversation with another professor at the university where I teach. She’s a professor of genetics. My background is in craniofacial genetics. I look at it as these people who have a genetic disease, we know the gene and the defective protein that causes it. Why are we not fixing the gene? Her response to me was that they had tried this in the past with kids and there were not great outcomes. It resulted in a few patient deaths. It was this blowback in the opposite direction. There’s this hesitance to use the technology until it was tried a million different times or there was some sort of social pressure that would influence that. Is that accurate? Have you heard about that at all?

Yeah. Part of it is a liability position. I’ve started talking with a number of doctors in America and it’s interesting. In America, you have places like Sloan Kettering, which is a top center. We’ve approached them. They’ve said, “When you have more data, we can offer it to patients.” They’re not going to start offering it. Mount Sinai can offer it because it was developed at Mount Sinai and they have a history with the technology.

I’ve started talking to a lot of private centers that are not necessarily affiliated with the top-known cancer centers. They have no problem. A lot of patients are going this route. Some of the oncologists are mapping pathways and saying, “This is your cancer mutation picture.” One oncologist that we work with says, “It’s your constellation,” which I love. It’s your personal constellation of mutations. He was like, “I’m going to hit this pathway with this drug.”

On some days, they’re treating patients with three cancer drugs. That’s much more toxic. It hasn’t been tested on an animal. They’re doing it in the air. It makes logistical sense but you haven’t modeled it in an animal. We are modeling it in an animal. The flies have always been used for toxicity screens because the flies are much more sensitive than us. We’ve already tested the combination and it’s already approved drugs. It’s not more than one cancer drug. It’s what some people are able to do but then we can be restricted as it’s crazy. I feel like in the US, there is a lot more say. If a patient wants something, there will be a road to get it. In the UK, it’s different.

Do you think that’s the climate of biotech in general? It’s so difficult for a small startup like yourself to make these end roads meet. One of the things that you highlighted was you would be better funded if you were a larger organization. Do you feel like the game is weighted against smaller startups like yourself?

We’ve probably taken on a very big problem. We’re not incrementally advancing in baby steps. We’re taking a big leap. That’s what it’s going to take to make a change but it is hard for a small company to make that big change on its own. There are other companies who are working towards the same. We’re all part of the same team. We’re not in competition but trying to make a change together.

We’re not incrementally advancing in baby steps. We’re taking a big leap in developing cancer treatments.

When I look at something like a COVID vaccine, it’s not technically a genetic therapeutic but it has some genetic basis to it because of the mRNA vaccine. That’s something that was despite a lot of negative blowback, it got out. Cancer is huge. There are other therapeutic options other than this one thing but we still don’t have a cure for it.

In comparison with the COVID vaccine to our technology, our technology is relatively safe because it’s combinations of approved drugs that are innocuous. We didn’t have a pandemic to have the response to it. Cancer is a pandemic. Cancer is increasing. It’s increasing in young people. I can’t tell you how many patients we have who are 20s, 30s, and 40s with young children. It’s heartbreaking. I lost my mother when she was in her 70s. I can’t imagine a child losing their mother in her 40s. It’s not fair. If these people want a chance at doing something that can give them a chance at life, it’s hard. I get angry about it. It makes me angry that these patients have a difficult time getting access.

One of the things that you highlighted is the younger demographic of patients who are coming in to see you and seek out cancer help in general. Do you feel like there are any trends that you can see with their types of cancer versus others? Is there more of a genetic component to it as opposed to the lifestyle choices that we hear about with smoking and things like that?

I don’t see it from that perspective because we look at the tumor and the blood. When we’re looking at colorectal cancer, people are presenting with it earlier. It’s something that advances so quickly. Most of the patients don’t know they have it until they’re in stage 3 or stage 4. They come to us and it’s difficult. I’ve only spoken about the personal discovery process that we have in which the patient comes, we model the tumor on the fly, and then we do drug screening.

We have a new technology we call TuMatch. TuMatch matches a patient but from whole exome sequence or whole genome sequence immediately to a treatment recommendation based on all the historical drug screening that we’ve done. For example, there are about 450 unique kinds of colorectal cancer by mutation profile. Let’s say 80% of the population will have 1 of 125 of these. We’ve modeled all 125 and started screening all of those models. That incoming patient will likely have an immediate match.

That’s helpful because it does take a long time to build a fly model and do the screening. It could take 6 to 7 months. A lot of patients were coming to us so they don’t have 6 to 7 months to wait for a recommendation. Unfortunately, a lot of our initial patients passed away before we were able to get the treatment. We call them pioneer patients. A lot of them knew that but they wanted to do it anyway because they wanted to pay it forward. They wanted in the future to be able to get immediate treatment.

My dad had anorectal melanoma. Are you familiar with that?

Yeah.

Is that on your screen of all the different ones that you’re looking at?

I’d have to look at the mutation profile because usually, melanoma, we don’t but rectal cancer, we do.

Anorectal melanoma is a melanoma of a non-kin exposed area. It’s in the GI tract. It’s universally fatal. He didn’t realize the diagnosis when he got it. He was like, “It’s a rectal cancer. I’m going to go through this path.” As soon as I got a diagnosis, my jaw was on the floor. The surgeon had told him about the overwhelming likelihood of mortality.

He still went through every single thing because he thought to himself the same way that you’re talking about. He was going to pay it forward. He was like, “Maybe some other person could get this.” He tried a few different clinical trials, none of which worked. He eventually succumbed to the disease. It’s interesting to see from that lens that there are these people who will volunteer themselves for unknown treatments. Some of these treatments were very difficult. Are you familiar with interferon therapy at all?

Yes.

I don’t know if your mom had it during her cancer experience. Interferon therapy was he was a different person. You could talk to him and he wouldn’t remember what you were saying. His personality changed. It was like a year that he was on this therapy and it did nothing for him. He was only doing it because he thought that there would be some sort of data that would be given to it to future generations.

That’s incredible. That’s admirable.

Thank you for saying that. My point is that’s an interesting thing. I feel like there are lots of people out there who would go through whatever therapy is available to them. Especially when you have an overwhelming likelihood of mortality, you only have a certain amount of time left. There’s a certain amount of population that’s like, “I’m not going to do anything.” That was my mom. My mom got to the point that when there was an overwhelming likelihood of mortality, she was like, “I’m done. I’m going to enjoy my life. We’re going to ride it out.” My dad was like, “I’ll do whatever. We will try whatever we can.” He always had that.

I feel like there is a certain subsection of the population. I don’t know if that’s something that you can tap into but there are patients out there who would be volunteering themselves because it’s a disease that cuts through every socioeconomic status and demographic. Have you seen patients like that? Have you had any experience with patients like that?

 

FSP-DFY 22 | Cancer Treatments

 

Most of our patients are exactly that patient. It’s $15,000. It is patients who have the means. We’ve created a foundation called the My Personal Therapeutics Foundation. We raise funds to support patients who could benefit from personalized therapy, which everyone can, and who can’t afford it. We’ve supported a few patients through this journey. We continue to raise funds to support new patients. The patients that can afford it are funding their journey when they know that their time is limited and it’s going to take time for us to get them a treatment.

Our patients are nice because if we have a patient that’s needing the therapy, we’ll put them ahead. There’s only so much screening we can do at a time. We have a line of fruit flies that need therapeutic screening. We can screen maybe 3 or 4 at a time. It usually goes in order of who pays but then, it goes in order of need. If a patient’s in remission or they started a new therapy and have six months until they’re going to start something new, we play with the line.

We have to let patients know because patients are like, “I was expecting a result this month.” We have to say, “There’s a patient who needs something.” They all care about each other’s position. They’re like, “Screen that person first. I’m good.” We’ve had this with all of the patients. Even the families, when a patient passes, a lot of the families still want to know the results. They either want to know that it’s published, shared, or entered our dataset and is going to help a new patient. We try to keep this community alive.

It is interesting how that mortality changes the way that people interact with each other. People become very gracious. It’s interesting. I did want to talk with you about the timeline. Let’s say somebody comes into you and wants your treatment. How long does it take for them to get those recommendations?

In the beginning, it was 6 to 7 months. For colorectal cancer, we have TuMatch. TuMatch is available for some pancreatic cancer and gastric cancer but mainly colorectal. As soon as we get a patient, if the patient has had sequencing before where they’ve had a Caris, a Foundation One, or a Tempus, we could sometimes use that. We’re developing too much as software and as a medical device. We’re going through the FDA and European regulatory agencies.

For it to be designated as a medical device, the input would be whole exome sequencing with a certain specification. It’s not like the Tempus and Caris. If a patient has that, we don’t want to have them encourage additional time. We’re not going to take a sound bolt and send it for additional sequencing unless we don’t see anything of interest in the Tempus or Caris report. Some patients say, “I’ve done the Tempus or the Caris. I still want the whole exome sequence. I have time so I’m going to do that.” It’s a month for the sequencing and then we get an immediate recommendation. That’s what we’re going to do for all the cancers.

At first, it was difficult for us because we are a small company. Patients come to us with different cancer types and we were taking them all. It is at the detriment of ourselves because for us, we’re building TuMatch and we want to screen all of these TuMatch avatars that we built. Patients have more complex tors where the avatars that we need to screen for TuMatch are more what we call building block avatars. It is 1 mutation, 1 plus 1 mutation, and 1 plus 3 mutations.

AI and machine learning like to have a dataset that’s not like that, not 40 unique profiles that aren’t related. We got a €2.5 million grant from the European Innovation Council to fund the development of TuMatch. We’re trying to take a little step away from patient screening. We’ll screen patients if they’re colorectal or pancreatic but we’re trying to stay away from other things.

We also work with BioPharma so we screen investigational drugs. We can help BioPharma understand how their drug works in patients with different molecular profiles and also identify combination partners for their drugs. We’re trying to move toward that element. Once we have enough contracts, then we’ll build a bigger facility. We could service more patients.

How much does the actual genome sequencing partake?

It takes about a month. We don’t do that internally. We could work with sequencing partners. In the US, we were working with Somagen. In Europe and the rest of the world, we’re working with Macrogen, as well as South Korea.

I’m still of the age where I can remember that the human genome being sequenced at all was a ten-year thing. We were jumping up and down as if we had landed on the moon or something like that. This time, it’s available to us in a month, which is pretty interesting.

The cost is going down too. Since we did the first whole genome sequencing years ago, the cost has gone down. Soon, it will be much cheaper and faster. What takes a bit longer and costs a bit more is that we do this tumor sequencing at a very deep depth because we want to resolve all of the subclones and understand what’s going on.

You could imagine that over the six months it would take us to identify a therapy, the patient’s tumor could evolve. What we found is that the mutations that are present in the beginning, if you have a deep enough view, you’ll see all of them. The same picture will still be the same in the clonal evolution. One might be stronger than another but it will still be the same bad guys. It becomes easy to understand when you’re doing a very deep read.

Have you had your genome sequence?

No, I haven’t. I’ve done 23andMe.

Do you have a special insight into your genome from 23andMe that someone like yourself would not have based on your experience?

I have a good understanding. I don’t have a lot of mutations that are indicative of future disease. One is macular degeneration. The other issues are eye-related.

I’m still a little hesitant. The biggest thing when I talk to people is they worry about the privacy issue and their genetic information being out there. For me, I don’t want to know. I want to live life.

I was scared. When I started Celmatix and we were developing the Fertilome test, we did the 23andMe as market research. We wanted to see how a patient feels and go through that journey. It was informative. I don’t care about the privacy component. I’d be more worried if people knew how broke I am and had my financial information.

I want to ask you as a side topic about the Fertilome test. My family members are trying that whole experience of trying to get pregnant past the age of normalcy. Was that more effective in getting outcomes of pregnancy? When you test somebody genetically, is that something that’s still out there? Is it therapeutic? It’s not something that I’ve heard about until you started.

It wasn’t a commercial success. It was scrapped, unfortunately. There are tests that are available and I can send you some. I wouldn’t endorse any publicly but there are tests that you can take. Ours was a bit more of an understanding of a woman’s egg quality and potential. It was also guiding them through whether they would be successful with their egg, whether they should try an egg donor, or if IVF wouldn’t work for them at all. It was more indicative of those things.

It’s interesting how that whole part of our society is going to play out. Women in general are having kids later and some are choosing not to have children. People are talking about all the population changes. I’ve never thought about the genetic component as being a factor until you said it so it was interesting to hear about that. Laura, thank you so much for being with us.

At the end of every session, I do have the three questions that I ask all of my guests so if you don’t mind indulging me. The biggest thing that I want to know is where you see genetics in ten years. Where do you hope that it’s going to be? Where do you realistically think that it’s going to be? Do you think that a lot of these promises that were talked about are going to materialize over the next decade or even twenty years from now, or do you feel like based on all of your understanding that might have to be tailored a little bit?

I’m very ambitious about our genetic future. My purpose of being and my purpose in life is transforming the way we look at genetic disease and personalizing medicine. I’m optimistic.

 

FSP-DFY 22 | Cancer Treatments

 

Another question I ask my guests is if you were to talk to somebody on the street, what is something in your field that you wish that everybody knew? That’s everybody from the person who is taking your order at a restaurant to the highest people in office. What would you say is something that you wish that everybody knew about, let’s say, genetics in general?

I’ll focus more on cancer. I wish that everyone understood how every cancer is different. If I had colorectal cancer and the person next to me had colorectal cancer, we have very different diseases and should be treated differently. Even a lot of doctors don’t understand that. Maybe they do understand it but they’re limited in what they’re able to offer. That’s important because changing that mindset will change a lot.

We have very different diseases and we should be treated differently

This is the last one. A lot of my interest in doing this show and being versed in cutting-edge technology, in general, comes from my interest in science fiction. It’s something that’s been a source of inspiration for me. I can look at some of the conversations that I’ve had. There are books that are particularly significant.

When you were talking about the whole privacy issue, I was thinking of the movie Gattaca, which I’m sure you’ve heard about considering your field. What are some sources of inspiration, whether it’s pop culture like movies and books or even other things that are a little bit more obscure that you draw inspiration that has brought you to this field?

I don’t look at it quite like that. I’m not so into science fiction. I’m into space. I’ve got a whole floor of my house that is devoted to the moon theme. I like stargazing and looking to the beyond in a real, literal way, not so much in a sci-fi way. Also, art in general. Artistry is an inspiration for me.

Thank you so much for being with us. Thank you to everybody who’s reading. We will see you again in the future. Have a great day.

Thank you so much.

 

Important Links

 

About Laura Towart

Laura Towart founded Vivan Therapeutics to advance a groundbreaking personalized approach to cancer developed at Mt Sinai Medical Center. Laura became passionate about personalized medicine during her PhD studies at Weill Cornell Graduate School for Medical Sciences/Memorial Sloan Kettering Cancer Center. Since then she has been actively involved in translational advances in medicine. In 2008 she co-founded Celmatix, and was the founding CEO, and helped build a next-generation women’s health company leveraging big-data and genomics. Laura is an avid startup and technology strategist and angel investor, with a focus on personalized medicine. Laura created the My Personal Therapeutics Foundation to provide financial support to patients who would like to access personalized cancer care but who cannot afford it. Laura is driven to help develop and foster biotechnology communities globally, and is an active advisor, speaker and mentor.

0 Comments

By: The Futurist Society