Quantcast
Channel: cancer Archives - VAI
Viewing all 60 articles
Browse latest View live

Connecting the genetic dots

$
0
0
Breast cancer is the most common cancer in women. Neurofibromatosis type 1 is a rare genetic disorder. A single gene may hold answers that could lead to new treatments for both.

An aggressive breast cancer and a rare disease marked by benign tumors on the nerves and skin have a common genetic root, one that may lead to improved therapies for both conditions, according to recent findings from Van Andel Research Institute scientists.

Dr. Carrie Graveel

“Rare diseases typically don’t receive as much attention because they affect a smaller number of people. Our findings are a great reminder of why it’s so important to study these disorders — the results can help people with rare diseases but also often have a much broader impact,” said Dr. Carrie Graveel, a VARI senior research scientist and breast cancer expert who led the project. “In this case, we found that NF1, the gene that causes neurofibromatosis, also greatly increases the risk of developing a particularly tough-to-treat type of breast cancer in women with and without neurofibromatosis.”

NF1 plays an important role in the body — it moderates cellular proliferation, ensuring that our tissues and organs aren’t overrun with a surplus of cells. When it malfunctions, cell reproduction spins out of control, resulting in either benign growths (such as those in neurofibromatosis) or in malignant tumors (such as cancer).

When NF1 was discovered and linked to neurofibromatosis in 1990, it was an exciting breakthrough in a disease that has largely thwarted attempts at treatment. Since then, scientists have continued combing the gene for clues in the hopes of translating their findings into new therapies that could help people around the world.

Their efforts are paying off: we now know that mutations in NF1 increase cancer risk (in fact, NF1 is one of the most commonly mutated or deleted genes in certain brain, lung and ovarian cancers).

And, even more recently, clinical evidence has suggested that NF1 elevates the risk of breast cancer, an effect that is even more pronounced in women with neurofibromatosis — below the age of 40, women with neurofibromatosis have a 10-fold increase in their risk of developing breast cancer.

Still, the fine details about the link between NF1 and breast cancer remained murky.

Dr. Matt Steensma

So Graveel and Dr. Matt Steensma, a physician-scientist at VARI and Spectrum Health and an authority on rare diseases, started digging into the data.

Together with their team, they scoured genetic information from METABRIC, a comprehensive compendium of nearly 2,000 breast cancer cases. Their findings were striking — a full quarter of cases in the database harbored a mutation in NF1, which also correlated to a marked decrease in survival.

Steensma and Graveel also found that NF1 is related to estrogen signaling in breast cancer and may play an important role in how breast cancers become estrogen-resistant. Estrogen is a critical hormone that also can fuel tumor growth.

But along with these new answers also come many new questions, Steensma said.

“Now that we’ve more firmly established the influence of NF1 on certain breast cancers, our next steps are to figure out ways to translate these findings into patient care,” he added. “There is still much more to discover that will help us on this path and, most importantly, that will help us find new, more powerful ways to help people with both of these diseases.”

This work was supported by the VARI Faculty Innovation Fund, the Breast Cancer Research Foundation and Tempting Tables.

The post Connecting the genetic dots appeared first on VAI.


High-throughput platform enables scientists to map activity of an emerging class of cancer drug targets

$
0
0

GRAND RAPIDS, Mich. (Nov. 28, 2018) — A powerful new biochemical platform is fueling the study of a family of enzymes that are promising targets for cancer treatment.

Dr. Scott Rothbart

Published today in Science Advances, the new method provides a high-resolution view of how these enzymes, called lysine methyltransferases, selectively mark proteins with chemical tags that alter their function. Because of their central role in all aspects of health and disease, proteins and the molecules that edit and interact with them often are targets for therapeutic development.

The platform was developed by Van Andel Research Institute’s Scott Rothbart, Ph.D., in collaboration with EpiCypher, Inc.

“This technology helps us to determine protein interaction networks for this understudied enzyme family based on chemical tagging,” said Rothbart. “Several inhibitors of these enzymes are currently in the clinical development pipeline for cancer therapy. Defining the spectrum of their activity is critical for understanding exactly how these drugs work and for selecting reliable biomarkers to track their activity in patients.”

A lysine methyltransferase attached to a substrate. The lighted portion depicts a methyl group being added by the methyltransferase.

Humans have approximately 20,000 genes that contain the instructions for making proteins, the molecular workhorses that are responsible for carrying out every process in the human body, from aiding in food digestion to managing communication between cells.

Once a protein is constructed, its function is often modified by the addition of small chemical tags, which instruct proteins where to go in the cell and when to perform their job. There are more than 100 different types of these tags, including the addition of methyl groups to the amino acid lysine.

Using their new technique, the team found that many more proteins may be tagged by lysine methylation than previously thought.

“Our study suggests that what we currently know about lysine methylation is just the tip of the iceberg,” said Evan Cornett, Ph.D., the study’s first author and a postdoctoral fellow in Rothbart’s laboratory at the Institute. “The method we developed will allow us to identify new targets across the full set of lysine methyltransferases in humans and, in doing so, help us and others determine which cancers and other diseases could benefit from treatments targeting this class of enzymes.”

Dr. Evan Cornett

This technology is the latest advance stemming from a collaboration between Rothbart’s lab and EpiCypher. Their work was supported by several National Institutes of Health (NIH) Small Business Innovation Research (SBIR) awards. Commonly known as America’s Seed Fund, SBIR provides federally funded research grants to small businesses in an effort to invest in American-led discovery. The SBIR program supports small businesses in the biotechnology sector, with a focus on strategies that have a high potential for significant impact and successful commercialization in the medical field. SBIR grants advocate for increased academic-industry partnerships to bridge the gap between basic science and clinical advancements, and are important stimulators of technological innovation.

“The beauty of this technology is its simplicity and throughput, which is staggering compared to current mass spectrometry-based approaches,” said Martis Cowles, Ph.D., EpiCypher’s Chief Business Officer and study co-author. “We are excited to use this technology to help drug developers identify new therapeutic targets and even identify optimal target substrates for high-throughput inhibitor screening.”

In addition to Rothbart, Cornett and Cowles, authors include Bradley M. Dickson, Ph.D, Robert M. Vaughan, Kevin M. Shaw and Philip P. Versluis, of Van Andel Research Institute; Krzysztof Krajewski, Ph.D., of University of North Carolina at Chapel Hill; Nicholas Spellmon, Ph.D., and Zhe Yang, Ph.D., of Wayne State University School of Medicine; Andrew Umstead and Irving E. Vega, Ph.D., of Michigan State University; Zu-Wen Sun, Ph.D., of EpiCypher; and Joseph Brunzelle, Ph.D., of Argonne National Laboratory.

Research reported in this publication was supported by Van Andel Research Institute and the National Institute of General Medical Sciences of the National Institutes of Health under award numbers R35GM124736 (Rothbart), R43GM110869 (Sun) and R44GM112234 (Sun). The content is solely the responsibility of the authors and does not necessarily represent the official view of the National Institutes of Health.

###

ABOUT VAN ANDEL RESEARCH INSTITUTE
Van Andel Institute (VAI) is an independent nonprofit biomedical research and science education organization committed to improving the health and enhancing the lives of current and future generations. Established by Jay and Betty Van Andel in 1996 in Grand Rapids, Michigan, VAI has grown into a premier research and educational institution that supports the work of more than 400 scientists, educators and staff. Van Andel Research Institute (VARI), VAI’s research division, is dedicated to determining the epigenetic, genetic, molecular and cellular origins of cancer, Parkinson’s and other diseases and translating those findings into effective therapies. The Institute’s scientists work in onsite laboratories and participate in collaborative partnerships that span the globe. Learn more by visiting vari.vai.org. 100% To Research, Discovery & Hope®

ABOUT EPICYPHER
A pioneer in the field of epigenetics and chromatin biology, EpiCypher® is a biotechnology company developing transformative technologies for researchers and drug developers worldwide. EpiCypher manufactures and sells a series of products and assay platforms that use recombinant “designer” modified nucleosomes (dNucs), including the SNAP-ChIP® product family for quantitative ChIP applications and the EpiDyne® product family for nucleosome remodeling assays, as well as recombinant histone binding proteins and enzymes, peptides and antibodies, and offers a broad range of custom substrate manufacturing and assay development services.

The post High-throughput platform enables scientists to map activity of an emerging class of cancer drug targets appeared first on VAI.

New combination blood test for pancreatic cancer may catch disease earlier

$
0
0

Dr. Brian Haab

GRAND RAPIDS, Mich. (Jan. 17, 2019) — A new approach to pancreatic cancer screening may help doctors detect the disease in people at high risk before it reaches more advanced and difficult-to treat stages.

A team led by Van Andel Research Institute (VARI) scientists has developed a new, simple blood test that, when combined with an existing test, detects nearly 70 percent of pancreatic cancers with a less than 5 percent false-positive rate. The results of the blinded study were published in Clinical Cancer Research, a journal of the American Association for Cancer Research.

Pancreatic cancer is difficult to diagnose because it often doesn’t have obvious early symptoms. By the time the disease is found, it typically is quite advanced, complicating treatment and leading to poorer outcomes. Only 8.5 percent of people with pancreatic cancer survive past five years, a figure that has risen just slightly since the early 1990s.

“Pancreatic cancer is an aggressive disease made even more devastating by its tendency to spread before detection, which is a serious roadblock to successful medical treatment,” said Brian Haab, Ph.D., a VARI professor and the study’s senior author. “We hope that our new test, when used in conjunction with the currently available test, will help doctors catch and treat pancreatic cancer in high-risk individuals before the disease has spread.”

Both tests detect and measure levels of sugars produced by pancreatic cancer cells that subsequently escape into the blood stream. The sugar measured by the new test — sTRA — is produced by a different subset of pancreatic cancers than CA19-9, the sugar measured by the existing test. When used together, the tests cast a broader net and detect subtypes of pancreatic cancer that may have been missed by using one of the two tests on its own.

The CA19-9 test was developed almost 40 years ago and detects only about 40 percent of pancreatic cancers. It currently is used to confirm diagnosis of pancreatic cancer or track disease progression rather than screen for the disease. The improved detection rate offered by the combined use of the sTRA and CA19-9 tests makes this approach a viable option for screening and early intervention, particularly in people who have a higher risk for developing the disease. This includes people who have a family history of pancreatic cancer, who have had pancreatic cysts or chronic pancreatitis, or who were diagnosed with type 2 diabetes later in life. Emerging evidence has suggested that sudden onset of diabetes after age 50 could be an early symptom of some pancreatic cancers. Currently, life-long diabetes is not considered to be a risk factor for or indicator of pancreatic cancer.

“We believe using these tests in a complementary fashion will help physicians detect pancreatic cancers much sooner in the disease process, which significantly improves a patient’s chance for survival,” Haab said. “Right now, there are few options for people suspected to have pancreatic cancer. This combined blood test could be a simple, cost-effective way to detect disease early enough to improve patient outcomes.”

Haab and his collaborators plan to partner with clinical laboratories to gain additional real-world validation for their method. If successful as expected, they envision the test becoming widely available as a screening tool for high-risk individuals.

In addition to Haab, authors include Ben Staal, Yin Liu, Ph.D., Daniel Barnett, Peter Hsueh, ChongFeng Gao, Ph.D., and Katie Partyka of VARI; Zonglin He, Ph.D., and Ying Huang, Ph.D., of Fred Hutchinson Cancer Research Center; Mark W. Hurd, Ph.D., and Anirban Maitra, MBBS, of MD Anderson Cancer Center; Aatur D. Singhi, M.D., Ph.D., and Randall E. Brand, M.D., of University of Pittsburgh; and Richard R. Drake, Ph.D., of Medical University of South Carolina. Barnett and Hsueh also are affiliated with Michigan State University. VARI’s Optical Imaging Core provided fluorescence imaging support.

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under award numbers U01CA152653 (Haab, Brand, Huang), U01CA200466 (Brand), U01CA200468 (Maitra), U01CA168896 (Haab, Brand, Huang), U01CA196403 (Maitra) and P30CA138313 (Drake). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

###

ABOUT VAN ANDEL RESEARCH INSTITUTE
Van Andel Institute (VAI) is an independent nonprofit biomedical research and science education organization committed to improving the health and enhancing the lives of current and future generations. Established by Jay and Betty Van Andel in 1996 in Grand Rapids, Michigan, VAI has grown into a premier research and educational institution that supports the work of more than 360 scientists, educators and staff. Van Andel Research Institute (VARI), VAI’s research division, is dedicated to determining the epigenetic, genetic, molecular and cellular origins of cancer, Parkinson’s and other diseases and translating those findings into effective therapies. The Institute’s scientists work in onsite laboratories and participate in collaborative partnerships that span the globe. Learn more about Van Andel Institute or donate by visiting www.vai.org. 100% To Research, Discovery & Hope®

Blog Post

Diagnosing a deadly cancer sooner and more definitively: An interview with Dr. Brian Haab

 

The post New combination blood test for pancreatic cancer may catch disease earlier appeared first on VAI.

Diagnosing a deadly cancer sooner and more definitively: An interview with Dr. Brian Haab

$
0
0

Pancreatic cancer is an insidious disease that often evades detection until it has spread beyond the pancreas, seriously complicating treatment and reducing the chances of successful remission. For years, there has been little progress toward finding improved ways to catch this deadly disease early on.

But that may be changing, thanks to a simple new combination blood test developed by Van Andel Research Institute’s Dr. Brian Haab and his colleagues. We asked Dr. Haab to tell us about the test, why early detection is so important and the next steps for moving the test into the doctor’s office.

Please tell us about your research and what you found.

Read Full Answer

We were looking for a blood test to detect pancreatic cancer based on the levels of specific molecules in the blood, where the levels, if they’re high, we would be able to detect pancreatic cancer versus some other benign condition of the pancreas.

The previous test — the best test so far — was developed 40 years ago. It’s not used for screening because it doesn’t detect enough pancreatic cancers with a less than 5 percent false-positive rate — it only gets less than half of the pancreatic cancers.

We have found a molecule that’s elevated in many of those patients that are low [in the molecule detected by the other test], so that by bringing the two together we can detect almost 70 percent of pancreatic cancer patients with a less than 5 percent false-positive rate. We’ve proven this now in blinded studies and we think we’re performing well enough that we can begin screening for the disease among people at a higher risk for developing pancreatic cancer.

What are the benefits of a blood test for pancreatic cancer?

Read Full Answer

A blood test is particularly valuable for pancreatic cancer because the symptoms are not visually evident like they are for some other cancers. A biopsy is difficult and potentially dangerous, imaging is expensive, it’s burdensome. A blood test is something that can be broadly applied and not very expensive.

How do you envision this test being used?

Read Full Answer

This test initially would be applied to people at a higher risk for developing pancreatic cancer, such individuals would be those with a family history of the disease, new-onset diabetes, obesity, a history of cysts or pancreatitis in pancreas. They would be at higher risk and so we would want to screen or monitor these patients to see if they are developing an incipient pancreatic cancer. If we can detect it early, they have a much better chance of having a good outcome.

Why are people who are diagnosed with type 2 diabetes later in life at a higher risk for pancreatic cancer?

Read Full Answer

The reason we screen people with new-onset diabetes is the new-onset diabetes may be the result of very early stage pancreatic cancer so they are at a higher risk of having a very early stage pancreatic cancer. If a person has had longstanding diabetes, they’re not at as high of risk for having early stage pancreatic cancer.

Why is it important to catch cancer early?

Read Full Answer

The reason we want to monitor for the early stage is because if the cancer can be detected at an early stage before it has spread throughout the organ or disseminated to other sites the outcomes for those patients are much better.

What are the next steps?

Read Full Answer

So, our goal is to make this available to patients and doctors. The next step in doing that is to get the assay established in a clinical laboratory that would be able to receive samples and run the test and give results back. We will initially get this available for our clinical partners on a research level and if we continue to see performance like we already have seen in our retrospective studies, we will offer if more broadly. We anticipate a relatively low-cost test that could have a high impact in detecting and diagnosing pancreatic cancer.

Read the news release here. Learn more about Dr. Haab’s work here.

The post Diagnosing a deadly cancer sooner and more definitively: An interview with Dr. Brian Haab appeared first on VAI.

10 things to know about pancreatic cancer

$
0
0

On March 12, the Institute will host A Focus on Pancreatic Cancer: From Foundations to Early Detection, a part of our Public Lecture Series, which is designed to engage and inform the community. The event is free and open, but registration is required (you can sign up here).

Attendees will hear from experts Dr. Brian Haab, whose research may lead to new ways to diagnose pancreatic cancer earlier and more definitively, and Dr. Bart Williams, whose work is shedding new light on how cancer cells communicate and how we may be able to block those signals, thereby treating cancer.

1) Pancreatic cancer is an aggressive, tough-to-treat disease in which malignant cells develop in the pancreas, a small gland located near the stomach that is responsible for producing hormones and digestive fluids. For more information, read our disease profile here.

2) Although the overall cancer death rate in the S. has dropped 27 percent in the last 25 years, pancreatic cancers continue to have a low survival rate — only about 8.5 percent of people with pancreatic cancer survive past five years.

3) Pancreatic cancer is the third leading cause of cancer death but is expected to surpass colorectal cancer as the second leading cause by 2020.

4) Risk factors for pancreatic cancer include:

  • Smoking
  • Obesity
  • A family history of pancreatic cancer or pancreatitis, a condition in which the pancreas is inflamed
  • A personal history of pancreatitis or chronic pancreatic cysts
  • Certain hereditary conditions (see here for a list)

5) In addition, emerging evidence has suggested that sudden onset of diabetes after age 50could be an early symptom of some pancreatic cancers. Currently, life-long diabetes is not considered to be a risk factor for or indicator of pancreatic cancer.

6) Not all pancreatic cancers are the same. First, they can arise from different cells within the pancreas — exocrine cells, which produce digestive fluids, and endocrine cells, which produce hormones. Roughly 95 percent of pancreatic cancers arise from exocrine cells.

That’s not the only difference. In 2017, scientists from The Cancer Genome Atlas reported results from in-depth analysis of 150 pancreatic cancer tumors, which confirmed major molecular factors that give rise to the disease and identified several new features that may one day lead to more precise, personalized therapies.

7) Pancreatic cancer is difficult to diagnose because it often doesn’t have obvious early symptoms (and those that it does have can have other, benign causes). By the time it is found, cancer cells may have spread throughout the organ or to other parts of the body — a process called metastasis — further complicating an already challenging treatment process and leading to poorer outcomes.

8) Sugars produced by pancreatic cancer cells act as a molecular fingerprint and may help catch these cancers early on. For example, Haab and his collaborators have developed a new, simple blood test that, when combined with an existing test, detects nearly 70 percent of pancreatic cancers with a less than 5 percent false-positive rate. Read more about the approach here.

9) Current treatments for pancreatic cancer are largely limited to chemotherapy and surgery, with effectiveness based in part on how widespread cancer cells are in the body (for more on treatment, please see the American Cancer Society’s pancreatic cancer resources here; all treatment decisions should be made in close consultation with an oncologist).

10) The more we know about pancreatic cancer, the better equipped we are to finds ways to catch it and treat it earlier, which could dramatically improve survival. Research, such as the work underway in the Haab and Williams labs, is moving us toward new strategies to give hope and more healthy years to people with pancreatic cancer.

Learn more about Dr. Haab’s research here and Dr. Williams’ research here.

The post 10 things to know about pancreatic cancer appeared first on VAI.

What is basic research and why is it important?

$
0
0

On March 12, the Institute will host A Focus on Pancreatic Cancer: From Foundations to Early Detection, a part of our Public Lecture Series, which is designed to engage and inform the community. The event is free and open, but registration is required (you can sign up here).

 Attendees will hear from experts Dr. Brian Haab, whose research may lead to new ways to detect pancreatic cancer earlier and more definitively, and Dr. Bart Williams, whose work is shedding new light on how cancer cells communicate and how we may be able to block those signals, thereby treating cancer. The lecture also will highlight the importance of basic research and how it fuels advances in the clinic. 

What is basic research?
Basic research seeks to answer fundamental questions about the world. It increases our knowledge and, in the case of biomedical research, lays the foundation for new treatment strategies for disease.

Questions that basic research seeks to answer include:

  • How do cells “talk” to each other?
  • How does a specific protein work?
  • How does the shape of a molecule affect its function?

For example, the lab of Dr. Bart Williams investigates a cellular communication network called Wnt (pronounced “wint”), which plays an important role in embryonic development, particularly in the formation of the bones and the heart. Problems with Wnt can result in an array of diseases, including breast cancer, glioblastoma (a type of brain cancer) and type II diabetes. Dr. Williams seeks to understand the nuts and bolts of Wnt down to the most minute level, solving a number of basic research questions while also setting the stage for impacting human health.

How does basic research impact health?
Basic research is the first step toward finding therapies for diseases like cancer and Parkinson’s. Once scientists understand the fundamental elements underlying our biology, they can leverage this knowledge to determine the causes of disease and find new treatments.

This process is called translational research because the scientist is translating basic research discoveries into applied solutions. In many ways, basic research is the springboard that propels health- and disease-focused research forward and, eventually, into the doctor’s office.

Translational research includes:

  • Figuring out how to switch off a gene that causes uncontrolled cancer cell growth
  • Understanding why bone is lost in osteoporosis and slowing or stopping the process
  • Determining why some cancers migrate to the bone and finding ways to prevent it

An example comes from the lab of Dr. Brian Haab, who developed a more precise method for diagnosing pancreatic cancer early on by detecting sugars in the blood produced by malignant cells. This approach, which combines a new test with a currently existing test, is now being investigated in a clinical setting and would not have been possible without a precise understanding of the basic biology of pancreatic cancer cells.

Learn more about Dr. Haab’s research here and Dr. Williams’ research here.

Looking for more information on basic and translational research? Check out our deep dive here.

The post What is basic research and why is it important? appeared first on VAI.

WATCH: Van Andel Institute Public Lecture Series: A Focus on Pancreatic Cancer

$
0
0

 

The first public lecture of 2019 at Van Andel Institute, A Focus on Pancreatic Cancer, from Foundations to Early Detection was held on March 12. Attendees learned why basic research is fundamental in scientists’ quest to discover more effective ways to detect and treat diseases like pancreatic cancer. They heard the latest news about a promising blood test that could help identify this aggressive disease much sooner than currently possible.

Pancreatic cancer poses a particular challenge to physicians and patients because it often doesn’t have obvious early symptoms. By the time the disease is found, it is typically quite advanced, complicating treatment and leading to poorer patient outcomes.

A Focus on Pancreatic Cancer highlighted new advances in disease detection and provided guests with an understanding of how laboratory research is changing our understanding of cancer.

  • This field is for validation purposes and should be left unchanged.

The post WATCH: Van Andel Institute Public Lecture Series: A Focus on Pancreatic Cancer appeared first on VAI.

Blocking epigenetic “Swiss army knife” may be a new strategy for treating colorectal cancer

$
0
0

GRAND RAPIDS, Mich. (April 4, 2019) — A new study out today in Cancer Cell shows that blocking specific regions of a protein called UHRF1 switches on hundreds of cancer-fighting genes, impairing colorectal cancer cells’ ability to grow and spread throughout the body.

Dr. Scott Rothbart

“Colorectal cancers are a growing public health problem, particularly in young people who haven’t yet reached the recommended age for regular screening,” said Scott Rothbart, Ph.D., an associate professor at Van Andel Research Institute (VARI) and co-corresponding author of the study. “UHRF1 has emerged in recent years as a key regulator of biological processes that drive colorectal cancer progression. This protein is akin to a molecular Swiss army knife, with many different parts that each have a different job. Revealing the parts that drive cancer progression could give us promising new ways to treat this disease.”

Colorectal cancers are the third leading cause of cancer-related death among both men and women in the U.S. While overall rates have been falling since the mid-1980s, the decrease is largely driven by routine screening in older adults. In people ages 20 to 39, rates are actually increasing — 1 to 2 percent a year between 1974 through 2013 for colon cancers and 3 percent per year between 1974 through 2013 for rectal cancers, according to a 2018 American Cancer Society report.

UHRF1 helps establish and maintain patterns of molecular tags on DNA, an epigenetic process known as methylation. These methylation patterns act like a switch that tells the body’s cellular machinery when specific genes should be turned on or off. In cancer cells, these patterns change, impeding the normal checks-and-balances on cell growth and allowing malignant cells to flourish and spread.

UHRF1 is a tantalizing new target that complements an emerging class of drugs called DNA methyltransferase inhibitors (DNMTis), which have shown exceptional promise for correcting methylation errors and treating cancer. However, many of the compounds developed to date have not been able to penetrate a sufficient number of tumor cells simultaneously to fully repair methylation and reactivate anti-cancer genes.

Dr. Stephen Baylin

“Epigenetic therapies are a promising avenue for cancer treatment, but we need new drugs that better reverse abnormal DNA methylation in patients’ tumors with fewer side effects,” said Stephen Baylin, M.D., co-corresponding author of the study, Director’s Scholar at VARI and Virginia and D.K. Ludwig Professor for Cancer Research at Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University. “The strong anti-cancer effects that result when we block the regions of UHRF1 responsible for maintaining methylation are a critical step toward developing new, improved inhibitors for cancer treatment”.

Identification of these regions on UHRF1 also may help better identify colorectal cancer subtypes, improving physician’s ability to take a personalized approach to treatment. Using data provided by The Cancer Genome Atlas, a National Institutes of Health-led effort to molecularly map cancer, the team found a strong correlation between high levels of UHRF1 and more aggressive, tough-to-treat malignancies.

They now hope to translate their findings into actionable prognostic markers and, eventually, improved therapies that are more tolerable to patients and more effective at fighting cancer.

In addition to Rothbart and Baylin, Limin Xia, Ph.D., of Tongji Hospital also is co-corresponding author.

Other authors include Xiangqian Kong, Ph.D., Wenbing Xie, Ph.D., Stephen M. Brown, Yi Cai, Ph.D., Srinivasan Yegnasubramanian, M.D., Ph.D., Yong Tao, Ph.D., Ray-Whay Chiu Yen, M.S., Michael J. Topper, Cynthia A. Zahnow, Ph.D., and Hariharan Easwaran, M.Sc., Ph.D., of Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Jie Chen, Kaichun Wu, M.D., Ph.D., Daiming Fan, M.D., Ph.D., and Yongzhan Nie, M.D., Ph.D., of State Key Laboratory  of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University; and Rochelle Tiedemann, Ph.D., of Van Andel Research Institute. Kong, Chen and Xie are co-first authors. Chen also is affiliated with Tongji Hospital.

Research reported in this publication was supported by National Institute of Environmental Health Sciences award no. R01ES011858 (Baylin), National Institute of General Medical Sciences award no. R35GM124736 (Rothbart), National Natural Science Foundation of China award no. 81522031 (Xia) and no. 81772623 (Xia), National Key Research and Development Program of China award no. 2018YFC1312103 (Xia), and the American Cancer SocietyMichigan Cancer Research Fund Postdoctoral Fellowship no. PF-16-245-01-DMC (Tiedemann). Research reported in this publication was also supported by The Hodson Trust (Baylin) and The Commonwealth Foundation (Baylin). The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

###

ABOUT VAN ANDEL INSTITUTE
Van Andel Institute (VAI) is an independent nonprofit biomedical research and science education organization committed to improving the health and enhancing the lives of current and future generations. Established by Jay and Betty Van Andel in 1996 in Grand Rapids, Michigan, VAI has grown into a premier research and educational institution that supports the work of more than 400 scientists, educators and staff. Van Andel Research Institute (VARI), VAI’s research division, is dedicated to determining the epigenetic, genetic, molecular and cellular origins of cancer, Parkinson’s and other diseases and translating those findings into effective therapies. The Institute’s scientists work in onsite laboratories and participate in collaborative partnerships that span the globe. Learn more about Van Andel Institute at vai.org.

Media Contact:
Beth Hinshaw Hall
Director of Communications & Marketing
Beth.HinshawHall@vai.org
616.234.5519

The post Blocking epigenetic “Swiss army knife” may be a new strategy for treating colorectal cancer appeared first on VAI.


Combating cancer from the lab to the clinic

$
0
0
May is Cancer Research Month, which raises awareness for the efforts underway to prevent, treat and, ultimately, cure cancer.

When it comes to combating cancer, collaboration is one of the most powerful tools in our arsenal. That’s why we teamed up with Stand Up To Cancer and other leading organizations, scientists and physicians four years ago to form the VARI–SU2C Epigenetics Dream Team — to see what we can do when our collective expertise and resources are combined.

The results to date are eight clinical trials at sites across the country and abroad, all aimed at developing new therapies that better fight cancer and that improve quality of life for people battling these diseases.

Here’s a snapshot of the cancers targeted by our trials:

Non-small cell lung cancer | 228,150 new cases each year (all lung cancers)
Non-small cell lung cancer is the most common type of lung cancer, and accounts for more than 80 percent of cases. Lung cancers are a major public health problem and claim more lives annually than any other type of cancer.

Acute myeloid leukemia (AML) | 21,450 new cases each year
AML is aggressive blood cancer that is notoriously difficult to treat and has poor long-term survival. It occurs when the bone marrow begins producing malignant red blood cells, platelets or myeloblasts (a special type of white blood cell).

Myelodysplastic syndromes (MDS) | 10,000 new cases a year (estimated)
MDS is a group of blood cancers marked by abnormal blood cells in the bone marrow. If left untreated, MDS can progress to AML, a more aggressive form of blood cancer. People who have previously undergone chemotherapy or radiation therapy often have an elevated risk of developing MDS.

Chronic myelomonocytic leukemia (CMML) | 1,100 new cases each year
CMML is a rare form of MDS in which the bone marrow produces too many myelomonocytes, a type of white blood cell, which then crowds out other important cells. If left untreated, CMML can progress to a more aggressive form of blood cancer.

Bladder cancer | 80,470 news cases each year
Bladder cancers are the sixth most common type of cancer diagnosed in the U.S. They are typically diagnosed in people older than 55, and occur more frequently in men than in women.

Pancreatic, liver, bile duct and gallbladder cancers | 56,770 new cases of pancreatic cancer each year; 20,000 new cases of liver and other biliary tract cancers each year
The biliary system comprises the pancreas, liver, gallbladder and bile ducts. Pancreatic cancer in particular poses a challenge to diagnosis and treatment; because it has few obvious early symptoms, it often is not caught until a more advanced stage, making it much more difficult to fight.

Colorectal cancer | 101,420 new cases of colon cancer each year; 44,180 new cases of rectal cancer each year
Colorectal cancer is the second leading cause of cancer death in men and women combined in the U.S. While the overall prevalence for the disease has steadily dropped since the 1980s, much of this decrease has been in older people, who frequently are screened for the disease. In younger people, the prevalence is rising and, since screening typically does not begin until age 50, these cancers often aren’t detected until a later stage when treatment is more challenging.

Hope for new and better treatments
These trials are a critical step on the road from the laboratory to the clinic and ensure the treatments being tested are safe and effective. If successful, the drug combinations being studied could help improve the lives of people suffering from these devastating diseases.

Learn more at www.vai.org/clinical-trials.

The post Combating cancer from the lab to the clinic appeared first on VAI.

Fighting cancer is an ever-evolving challenge

$
0
0
This year’s Origins of Cancer symposium explores cancer evolution, from genetics to selection pressure

By Guillermo Flores and Leslie Wyman
Van Andel Institute Graduate School Ph.D. students and organizers of the 2019 Origins of Cancer symposium

Why have we not yet cured cancer?

As scientists, we get this question a lot.

After all, we read about new discoveries every day in scientific journals and in the news. Moreover, we know that cancer is largely a genetic disease. Scientists have already sequenced the entire human genome; it should be as easy as finding the differences between tumor cells and healthy cells and then just fixing the problems, right? Or perhaps we could exploit these differences to come up with an effective, individual treatment strategy for each patient — the pinnacle of true, personalized therapy.

Yet despite these advances and humanity’s collective efforts, cancer persists.

Worse still, cancer sometimes appears to be cured only to return with a vengeance. Countless hours of work from some of the world’s brightest minds, incalculable amounts of money, innumerable tragic stories of those touched by cancer and an ever-increasing awareness have certainly yielded positive results.

Yet, cancer persists.

That brings us back to the question — why haven’t we cured cancer?

To find the answer, we have to recognize one crucial fact — cancer evolves. It does so in a fashion not so different from what Charles Darwin described 150 years ago in his groundbreaking book On the Origin of Species. Cancer is subject to natural selection just like organisms and viruses. Unfortunately, when it comes to survival of the fittest, cancer cells use natural selection to become hardier and tougher to treat.

If we want to finally cure cancer, we need to understand how and why it evolves. That’s the focus of this year’s Origins of Cancer, a one-day scientific symposium that will be held July 26 at Van Andel Institute.

Scientists have begun to unravel questions into how complex genetic machinery orchestrates natural selection on a cellular level and how those processes contribute to human health and disease. Intense research is underway to understand the mechanisms of how cancer begins, changes with time and becomes resistant to therapy. The 2019 Origins of Cancer will explore these topics, specifically:

We are also honored to host Prof. Charles Swanton as our keynote speaker. Prof. Swanton combines laboratory research at the Francis Crick Institute with clinical duties at University College London Cancer Institute. His research focuses on how tumors evolve over space and time. Prof. Swanton has helped to define the branched evolutionary histories of solid tumors, processes that drive cancer cell-to-cell variation in the form of new cancer mutations or chromosomal instabilities, and the impact of such cancer diversity on effective immune surveillance and clinical outcome.

Origins of Cancer will be held at the Institute July 26. For more information and to register, visit originsofcancer.org.

Variation: In genetics, variation refers to DNA differences between individuals. Even identical twins have slight differences in their DNA. There is typically significant variation between healthy cells and cancer cells.

Inheritance: Before a person is born, the DNA from each biological parent is first shuffled in the sperm and egg. During fertilization, two sets of shuffled DNA are combined. All this adds up to increased variation. Cancer cells often mutate. By dividing, new cancer cells can inherit these mutations.

Selection: Often referred to as “survival of the fittest,” selection is the mechanism by which evolution occurs. If a gene increases the chance for an individual to survive it will become more common in a population over time because individuals with that gene are more likely to survive and pass it down to their offspring. Conversely, if a gene decreases the chance for an individual to survive, it will become less common because it is less likely to be passed on to future generations.

The environment plays a big part by applying what is referred to as a “pressure.” If the stakes of living and dying are raised, there is an increased pressure and selection becomes more powerful. For example, if there is a gene that makes a cancer cell resistant to chemotherapy, it may be just as common as any other gene until chemotherapy is applied. Once the pressure of chemotherapy is present the gene becomes much more important for survival and “selects” for these resistant cells to survive. Alas, this is commonly the basis of tumor recurrence and relapse.

The post Fighting cancer is an ever-evolving challenge appeared first on VAI.

10 research trends we’re excited to watch in the next decade

$
0
0

With a new decade comes a new world of possibility and innovation waiting to be unleashed in labs at Van Andel Institute and across the globe. As we take our first steps into the 2020s, here are 10 promising research trends to keep an eye on in the coming years.

Our understanding of Parkinson’s disease will continue to evolve, spurring new research and breakthroughs

Parkinson’s disease has long been considered a movement-related disorder that originates in the brain. Recent research, however, suggests that Parkinson’s disease might represent a group of closely related disorders. While they share key clinical features, they may have roots in different anatomical locations, such as the gut and nose. Even if the anatomical starting points differ, there is evidence that cell metabolism, inflammation and poor handling of certain proteins are common features. In the coming years, these insights could lead to the development of new therapies to slow or stop its progression.

Breakthroughs in common diseases like Parkinson’s, dementia with Lewy bodies and Alzheimer’s might help us better understand rare disorders

It’s become increasingly clear that an improved understanding of one disease may provide new insights into another. For rare and understudied diseases like multiple system atrophy (MSA), this provides a particularly important opportunity to gain the insight required to develop new diagnostics and treatments.

Inflammation will increasingly be linked to disease — and give rise to new treatments

Inflammation is a normal part of the immune system, responsible for marshalling resources to the site of a wound, infection or disease (like cancer) to help the body fight back. Yet when inflammation remains longer than needed, it can cause or contribute to a host of health problems. Researchers are hard at work sorting out exactly how and why this happens, findings that could serve as the foundation for new therapies for cancer, neurodegenerative diseases, depression and many others.

Understanding how the body processes, uses and stores energy will be critically important

Metabolism powers every aspect of the human body, from keeping the immune system running to ensuring our hearts have enough energy to beat. We’ve known for some time that metabolic dysfunction plays a central role in diseases like diabetes and even in cancer, thanks to malignant cells’ voracious appetites for energy. But new breakthroughs also have linked issues with metabolism to neurodegenerative diseases like Parkinson’s and Alzheimer’s, among others. As we better understand our bodies’ incredibly complex web of metabolic processes, it’s likely that we will continue to find metabolism at the center of many disorders — and many future treatments.

Cancer treatment will harness combinations of medications designed to give cancer a one-two punch

We now know that cancer comprises more than 100 different diseases, each with its own litany of subtypes (for example, breast cancers can be defined based on the presence or absence of certain molecular receptors). This expansive diversity means that there is likely no silver bullet cure. Instead, we can expect to see more combination therapies that utilize multiple medications whose effects complement or enhance each other’s ability to fight cancers. A prime example is pairing an immunotherapy drug, which bolsters the body’s natural defenses against cancer, with an epigenetic drug that makes cancer cells more recognizable, more susceptible to immune attack and is thought to reinvigorate exhausted immune cells and get them back in the fight.

We will better understand how the health and diet of one generation can affect the next

Can a parent’s diet, experiences and lifestyle impact their children? Or subsequent generations? Mounting evidence suggests that nutrition does indeed have a ripple effect across generations, even altering individuals’ predisposition to disease. Efforts are underway to better understand how nutrition and other environmental factors reprogram the genome and epigenome, and how this information traverses generations. The goal?  Developing ways to prevent and treat diseases including diabetes, cancer, and Parkinson’s, and to protect our children from the unwanted consequences of our own lifestyles.

We will see the translation of structural findings into actionable therapies

Structural biology seeks to determine shape and architecture of life’s smallest building blocks, such as proteins. These efforts are vitally important, helping us understand in intricate chemical detail how the body works and laying the earliest foundations for new medications. In the past five years, there has been a boom in structural research, based on technological advances in cryo-electron microscopy (cryo-EM), a technique that helps scientists view molecules down to the atomic level. In the next decade, we will continue to build on these foundational discoveries, which could give rise to new treatments.

We’ll learn more about microenvironments and microbiomes and their role in cancer and other diseases

In many ways, the future will be “micro.” Our technology is miniaturizing, we are able to study the tiniest components of life in ways never before possible and scientists are increasingly understanding the importance and diversity of microenvironments, the mini ecosystems that exist throughout the body. In cancer, the microenvironment is the area immediately surrounding a tumor, which can comprise blood vessels, immune cells and structural cells, among others. These areas offer windows into how specific cancers grow and proliferate, and may even offer new solutions for stopping them. We’re also learning more about the role of microbiomes, the host of microscopic organisms such as bacteria that inhabit specific areas of the body like the gut. For example, researchers who are sleuthing out the links between the gut and Parkinson’s disease are taking a look at the gut microbiome as a possible factor in disease onset.

Technology will to spur even more innovation

Editing genes using CRISPR. Determining molecular structures with high-powered cryo-EM. Decoding the genome through next-generation sequencing. The advent of these powerful technological tools, paired with increasingly robust computational capabilities, have revolutionized how we study health and disease, and already have contributed to critical biomedical breakthroughs. With technology continually advancing, we can only expect the innovation to continue in years to come.

Big Data will continue to yield new insights

Big Data refers to the massive amounts of information generated by certain types of research, such as genomics. With technology improving every day, the amount of data being churned out will only continue to balloon, presenting opportunities and challenges to scientists working to get to the root of disease. By closely analyzing large datasets (a monumental task itself), scientists can look for patterns linked to disease. A prime example is The Cancer Genome Atlas, a National Institutes of Health-led effort that molecularly mapped 33 different types of cancer. These highly detailed and vast datasets helped identify important variations in cancer types and subtypes that may form the foundation of new targeted therapies.

The post 10 research trends we’re excited to watch in the next decade appeared first on VAI.

World Cancer Day 2020: What energizes VAI scientists about the future of cancer research

$
0
0

February is National Cancer Prevention Month, and Feb. 4, 2020, is World Cancer Day.

To mark the occasion, Van Andel Institute scientists shared what most excites them about the directions cancer research will head in the next decade. Their insights range from personalized medicine to better harnessing the power of data ecosystems and informatics tools.

For more information on VAI’s cancer research, click here.

Peter A. Jones, Ph.D., D.Sc. (hon)
VAI Chief Scientific Officer
Distinguished Professor, Center for Epigenetics

“The last decade of cancer research has revolutionized our understanding of these diseases, given rise to life-changing new treatments and laid the groundwork for the next generation of cancer care. As we go forward, I am most excited for the potential of these advances to fuel development of even better therapies and prevention methods to further reduce cancer mortality, giving people more healthy years.”

Andrew Pospisilik, Ph.D.
Director and Professor, Center for Epigenetics
Member, Metabolic and Nutritional Programming, Center for Cancer and Cell Biology

“I am most excited about the power of numbers. Big data and sequencing are yielding insights at an increased rate. We are still slow to harness the power and energy within the big data ecosystem. In the next decade, the continued advance of informatic tool development will enable researchers to reach unbiased insights with greater speed, power and accuracy than ever before.”

Peter Laird, Ph.D.
Professor, Center for Epigenetics

“I look forward to the day when personalized targeted therapy, based upon comprehensive genomic and epigenomic analysis, becomes the standard of care in clinical practice, offering far greater precision and efficacy in our cancer care.”

Gerd Pfeifer, Ph.D.
Professor, Center for Epigenetics

“While we continue to improve cancer therapy, the biggest goal should be to prevent cancer in the first place. However, to prevent cancer, we need to understand what causes it and how that works. I expect that the next decade will see new insights into the mechanisms of cancer causation, which will differ according to the tissues in which the tumors arise.”

Xiaobing Shi, Ph.D.
Professor, Center for Epigenetics

“For what I can imagine for now, the most exciting thing to look forward to cancer research (and treatment) in the next decade is to bring artificial intelligence into our radar. AI has started to show influence on many aspects of our life: automotive, video games and more. I expect AI will do a lot more to improve human health in the coming decades, not only for clinical diagnosis, prevention and patient treatment, but also in cancer research in labs. Exciting!”

Hong Wen, Ph.D.
Associate Professor, Center for Epigenetics

“Great progress has been made in large-scale genomics-based tumor evaluation, liquid biopsy and immunotherapy in recent years, and the momentum will continue in the next decade. We are at a very exciting time to witness how progress in these research fields and information obtained will affect the development of personalized precision medicine for cancer treatment. This research will also help to reduce side-effects from unnecessary cancer treatment and benefit cancer prevention.”

Brian Haab, Ph.D.
Professor, Metabolic and Nutritional Programming, Center for Cancer and Cell Biology
Assistant Dean, Van Andel Institute Graduate School

“What most excites me are metabolic and nutritional approaches to treating cancer, and the ability to determine the subtypes that will respond to each treatment. The ability to detect, diagnose, and determine the subtype of cancers more precisely efficiently may lead to better treatments and outcomes.”

Xiaohong Li, Ph.D.
Assistant Professor, Skeletal Disease and Cancer Therapeutics
Center for Cancer and Cell Biology

“Metastases account for 90% of cancer deaths. Circulating tumor cells (CTC, or cancer cells found in a patient’s blood) and disseminated tumor cells (DTC, or cancer cells found in a patient’s bone marrow) are detected as early as when patients are first diagnosed with cancer. These cells are believed to be the cells of origin of metastases and recurrence. The more we learn about these cells, the more we will provide better diagnosis, prognosis and treatment for metastases and recurrence.”

The post World Cancer Day 2020: What energizes VAI scientists about the future of cancer research appeared first on VAI.

Explainer: What are mitochondria?

$
0
0

If you ask high school biology students about mitochondria, they’re likely to respond with the same answer: mitochondria are the powerhouses of the cell.

But let’s get more specific. To start, mitochondria are organelles (tiny cellular versions of organs) that generate most of the energy needed for cells to function and survive.

Mitochondria march to the beat of their own drum

Unlike other parts of a cell, replication of these power-plant organelles is not tied to cell division — mitochondria divide independently of the cell that they call home. This, among other unique features, are holdovers from ancient ancestors of mitochondria, which were likely free-living, single-celled organisms called prokaryotes.

The thinking goes that an ancient symbiosis resulted when a cell containing a nucleus (the “command centers” of certain cells) engulfed a prokaryote, which does not have its own nucleus. The host cell came to rely on the prokaryote for energy production, while the prokaryote used the host cell for protection. Over time, this relationship developed into what we see today: a cell that relies on its mitochondria for energy.

Mitochondria also have their own very small and widely varied genome — another trait carried over from their single-celled ancestors — and are generally inherited only from the mother.

How can we use our understanding of mitochondria to battle disease?

When mitochondria are not functioning properly, problems can arise that then contribute to disease — including cancers or neurodegenerative diseases like Parkinson’s.

Take cancer cells, for example, which have voracious appetites for energy. If we can find ways to turn down or stop these mitochondrial power-plants in malignant cells, we may be able to deprive the cells of the energy required to spread and, ultimately, kill them (read more about a recent study on that topic here).

Evidence also suggests that mitochondria have a role to play in the onset and progression of Parkinson’s disease, a neurodegenerative disorder for which there currently are no treatments that slow or stop progression. A wealth of recent research suggests that a breakdown in brain cells’ ability to produce energy may interfere with their “housekeeping” systems, allowing abnormal, toxic proteins to pile up, eventually killing these critical cells. The death of these cells leads to Parkinson’s hallmark symptoms, such as loss of the ability to move.

What does all of this mean for treating disease?

Many medications already exist that affect cellular energy produced by mitochondria, such as those used to treat diabetes. In fact, several medications for Type 2 diabetes are being investigated as potential therapies for cancer and Parkinson’s (read more about these efforts in cancer here and in Parkinson’s here).

The post Explainer: What are mitochondria? appeared first on VAI.

Van Andel Institute scientists win Team Science Awards from American Association for Cancer Research

$
0
0

Peter Laird, Ph.D.

GRAND RAPIDS, Mich. (June 22, 2020) — The American Association for Cancer Research has awarded Van Andel Institute Professor Peter W. Laird, Ph.D., Director’s Scholar Stephen B. Baylin, M.D., FAACR, and Associate Professor Hui Shen, Ph.D., 2020 AACR Team Science Awards for their pivotal roles in the establishment and success of The Cancer Genome Atlas (TCGA), a landmark project that revolutionized our understanding of cancer and that is hailed as an exemplar of scientific collaboration.

The first Team Science Award includes Laird and Baylin as well as other project leaders and individuals who were central to TCGA. The second Team Science Award includes Laird, Baylin and Shen, along with 127 additional members of the current TCGA project team. Awardees will be honored during a special session of the AACR Virtual Annual Meeting II on Wednesday, June 24.

Stephen Baylin, M.D.

“The insights revealed by TCGA will continue to transform cancer research and treatment for years to come. We are thrilled to join AACR and the scientific community in celebrating the contributions of Dr. Laird, Dr. Baylin, Dr. Shen and all those who took part in the project,” said Peter A. Jones, Ph.D., D.Sc. (hon), VAI Chief Scientific Officer. “TCGA truly is a testament to the power of team science.”

Announced in 2005 and completed in 2018, TCGA was a National Cancer Institute and National Human Genome Research Institute-led effort to molecularly map 33 cancer types, including 10 rare cancers.

In all, hundreds of scientists in the U.S. and Canada participated, resulting in detailed analyses of more than 20,000 biospecimens and numerous scientific publications that continue to fuel new breakthroughs and innovations. It has become an invaluable asset for the development of precision therapies and the identification of new drug targets.

Hui Shen, Ph.D.

Following TCGA’s inception, Laird and Baylin established and co-led the epigenetics analyses arm of the project, which cataloged molecular changes that influence cancer risk by altering how genes are expressed rather than the genes themselves. Laird went on to lead epigenetic analyses for TCGA through its completion, as well as the project’s effort to characterize cancers by their molecular subtype.

Laird began working on TCGA while serving as a professor at University of Southern California, and continued when he joined VAI in 2014.

Shen, who also joined VAI from USC in 2014, played a major role in epigenetic data analyses for the project.

Baylin, who co-leads the Van Andel Institute–Stand Up To Cancer Epigenetics Dream Team, holds a primary appointment at Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University. His insight and guidance were central to the genesis and growth of the epigenetics component of TCGA.

“TCGA was a vastly complex undertaking that demonstrated the power of team science initiatives,” Laird said. “Each person who worked on the project played a part in its success and in charting future cancer research and therapies built on its findings.”

AACR’s announcement is available here.

###

ABOUT VAN ANDEL INSTITUTE
Van Andel Institute (VAI) is committed to improving the health and enhancing the lives of current and future generations through cutting edge biomedical research and innovative educational offerings. Established in Grand Rapids, Michigan, in 1996 by the Van Andel family, VAI is now home to more than 400 scientists, educators and support staff, who work with a growing number of national and international collaborators to foster discovery. The Institute’s scientists study the origins of cancer, Parkinson’s and other diseases and translate their findings into breakthrough prevention and treatment strategies. Our educators develop inquiry-based approaches for K-12 education to help students and teachers prepare the next generation of problem-solvers, while our Graduate School offers a rigorous, research-intensive Ph.D. program in molecular and cellular biology. Learn more at vai.org.

The post Van Andel Institute scientists win Team Science Awards from American Association for Cancer Research appeared first on VAI.

Explainer: How do gene mutations happen?

$
0
0

Many diseases and disorders studied at Van Andel Institute have genetic links, including cancer and Parkinson’s disease. Gene mutations, or changes to genes, play a major role in disease. Here’s a quick primer.

What are genes?

Courtesy of the Genetics Home Reference, National Institutes of Health

Before we get too deep, let’s cover the basics. Most human cells have 23 pairs of chromosomes, for a total of 46 individual chromosomes. We inherit our chromosomes from our parents — one from our mother, and one from our father. Each chromosome consists of tightly coiled DNA (deoxyribonucleic acid), a ladder-like molecule that carries all of the instructions needed for life. The “rungs” of DNA’s ladder comprise pairs of four chemicals — adenine and thymine, and cytosine and guanine. Different patterns of these chemical pairs result in instructions for making everything your body needs for you to be you.

Genes are segments of DNA that determine specific human characteristics such as height, eye color or the risk of developing a certain disease. Most genes are the same in all people, but a small number of them (less than 1% of the total) differ slightly between individuals. These alleles — the forms of the same genes with small differences in the sequence of their DNA bases, or the chemical “rungs” of the ladder — contribute to each person’s unique features.

Some characteristics arise from a single gene, while others result from gene combinations. The Human Genome Project estimated that humans have between 20,000 and 25,000 genes, and every person has two copies of each gene — one inherited from each biological parent.

Epigenetics, or shifts in how our genetics are regulated, play a role in our understanding of genes and their expression, too. Read more about it here.

What is a gene mutation?

A gene mutation is a permanent change in the DNA sequence that makes up a gene. When this occurs, we’re left with a sequence that differs from what is found in most people. Mutations can affect a single DNA base pair to a large segment of a chromosome that includes multiple genes.

Gene mutations can be classified in two major ways:

  1. Hereditary mutations are inherited from a parent and are present throughout a person’s life in virtually every cell in the body. Also known as germline mutations, these changes come from the parent’s egg or sperm cells; when these germ cells unite, the resulting fertilized egg cell gets its DNA from both parents. If this DNA has a mutation, the offspring will have the mutation in each of their cells.
  2. Acquired mutations are the opposite. They occur after the fertilization of an egg cell and can be caused by environmental factors such as ultraviolet radiation from the sun or errors made as DNA copies itself during cell division. These are also the most common cause of cancer (germline mutations account for only about 5% to 20% of all cancers).

Acquired mutations in cells other than sperm and egg cells cannot be passed to the next generation and are known as somatic mutations.

Are all mutations harmful?

No, not all mutations are harmful. Most disease-causing gene mutations are relatively uncommon in the general population.

Take for example polymorphisms, which are variations of a particular genetic sequence. Common enough to be considered a normal variation in DNA, polymorphisms result in many of the normal differences between people such as eye color, hair color and blood type. Some polymorphisms involve a single change at a base pair (DNA’s “rungs”) while others are much bigger.

Though many polymorphisms don’t negatively affect a person’s health, some variations may influence the risk of developing certain disorders and can serve as biological markers that may help scientists locate genes associated with specific diseases.

How can we use our understanding of gene mutations to battle disease?

When we know what a specific gene or a mutation does, we can identify when something has gone awry and work to develop ways to fix the problem. Finding certain mutations in cells helps confirm diagnoses of specific cancers, and can be used after diagnosis to determine the therapies to which the cancer will best respond.

By understanding the genetic components and gene mutations associated with specific diseases, VAI scientists can translate their discoveries into actionable prevention and treatment techniques to help people live longer, healthier lives. A few recent examples include:

  • The work being done in the Moore Laboratory to uncover the genetic causes of Parkinson’s disease, with the goal of developing targeted therapies for the disease.
  • Research related to genome-wide association studies (GWAS) conducted in VAI’s Coetzee Laboratory with the goal of understanding how the genetic predisposition of complex diseases such as cancer and Parkinson’s impose risk to aid in the development of therapies that slow or halt the diseases.

Work related to The Cancer Genome Atlas (TCGA), a landmark project that revolutionized the understanding of cancer and that is hailed as an exemplar of scientific collaboration. The more than decade-long initiative, led by the National Institutes of Health, was the most in-depth undertaking of its kind, and included analysis of 10,000 tumors across 33 cancer types.

The post Explainer: How do gene mutations happen? appeared first on VAI.


Van Andel Institute to serve as Cancer MoonshotSM Biorepository

$
0
0

GRAND RAPIDS, Mich. (September 15, 2020) — Van Andel Institute’s Biorepository has been awarded a $2.7 million, two-year subcontract from the Frederick National Laboratory for Cancer Research currently operated by Leidos Biomedical Research, Inc. on behalf of the National Cancer Institute to serve as the biorepository for the Cancer Moonshot Biobank study, a national initiative to transform cancer treatment and prevention through accelerated research.

In this role, VAI will assemble and distribute kits for the collection of tumor tissue, blood and other biospecimens to hospitals and medical centers around the U.S. Once samples are collected from volunteers, they will be shipped to VAI for processing and either stored for later study or sent to other organizations conducting analyses for the Cancer Moonshot. In all, the Biobank project is expected to collect biospecimens from more than 1,000 participants.

“Biospecimens are the bedrock of scientific research — without them, we wouldn’t be able to study cancer or develop new treatments and diagnostics,” said Scott Jewell, Ph.D., director of VAI’s Core Technologies and Services, which includes the Institute’s Biorepository. “We are honored to be part of the Cancer Moonshot Biobank study and look forward to doing our part to support research and improve cancer care.”

The Cancer Moonshot was launched in 2016 by the Obama Administration. Its strategic aims, determined by a Blue Ribbon Panel of experts, are designed to answer critical scientific and medical questions while ensuring the samples collected represent the diversity of the U.S. population.

VAI’s Biorepository provides services for a number of large-scale national and international projects, including NIH’s Clinical Proteomic Tumor Analysis Consortium and the National Cancer Institute’s Biospecimen Pre-Analytical Variables Program. The Biorepository team also played an integral role in biospecimen collection for the NIH’s Genotype-Tissue Expression project, including developing and shipping the kits used by investigators around the country to collect tissue samples. It currently serves as the biobank for the Multiple Myeloma Research Foundation’s CoMMpass Study, the Tuberous Sclerosis Alliance and the Van Andel Institute–Stand Up To Cancer Epigenetics Dream Team. Since 2012, VAI’s Biorepository has been accredited by the College of American Pathologists (no. 8017856), which provides objective assurance that VAI meets or exceeds the high standards set by CAP.

The project has been funded in whole or in part with federal funds from the National Cancer Institute of the National Institutes of Health under contract no. HHSN261201500003I, Task Order HHSN26100042 through Leidos Biomedical Research, Inc. under subcontract no. 20X062Q. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the U.S. government.

Media Contact
Beth Hinshaw Hall
Van Andel Institute
Beth.HinshawHall@vai.org
616-822-2064

###

ABOUT VAN ANDEL INSTITUTE
Van Andel Institute (VAI) is committed to improving the health and enhancing the lives of current and future generations through cutting edge biomedical research and innovative educational offerings. Established in Grand Rapids, Michigan, in 1996 by the Van Andel family, VAI is now home to more than 400 scientists, educators and support staff, who work with a growing number of national and international collaborators to foster discovery. The Institute’s scientists study the origins of cancer, Parkinson’s and other diseases and translate their findings into breakthrough prevention and treatment strategies. Our educators develop inquiry-based approaches for K-12 education to help students and teachers prepare the next generation of problem-solvers, while our Graduate School offers a rigorous, research-intensive Ph.D. program in molecular and cellular biology. Learn more at vai.org.

The post Van Andel Institute to serve as Cancer MoonshotSM Biorepository appeared first on VAI.

Graduate student spotlight: Investigating a cancer linked to a rare genetic disorder

$
0
0

Throughout the year, we highlight Van Andel Institute Graduate School’s doctoral students. This month features Lauren McGee, a student in the lab of Dr. Matt Steensma. Lauren studies a tough-to-treat cancer linked to a rare genetic disorder in hopes of developing new therapies.

Q: What do you study?

LM: I work on a cancer that arises from a genetic disorder called Neurofibromatosis Type 1, which causes dysregulation of an important cell growth pathway. This cancer, called malignant peripheral nerve sheath tumor (MPNST), has no chemotherapeutic options and quickly develops resistance to any initial treatments that might seem to work at first. The goal of my thesis project is to understand how this dysregulation leads to the development of treatment resistance, and to identify new pathways that can be targeted to improve the outcome of patients with MPNST. 

Lauren McGee

Q: Did you take time off before starting your Ph.D. degree or come directly from an undergraduate or master’s degree program?

LM: I was a post-baccalaureate fellow and then a contract scientist at the National Center for Advancing Translational Science (NCATS) at the National Institutes of Health in Rockville, Maryland, for three years between my undergrad and starting at Van Andel Institute. I’m incredibly grateful for that time; I worked with amazing people and I learned how to be a full-time scientist, which has really helped me be successful in graduate school.

Q: Why did you choose Van Andel Institute Graduate School?

LM: I really enjoyed the small feel of the Institute, and the small size of the graduate program specifically. I think having these smaller-sized classes helps foster a community within the graduate program and ensures that no one is really a stranger here. The problem-based learning was another draw, especially taking time off to work in the “real world” per se; this approach felt very similar to how I was learning about my projects at the NIH, and in general felt like a more natural progression of knowledge and understanding of the material. I also liked that students were supported by the graduate program and not just the principal investigators (PI). Other places I interviewed warned that the PI you might want to work with may not have funding, so you would have to find another lab to join. That isn’t an issue here, which eased a lot of stress.

Q: What is your favorite stress-reduction technique?

LM: My favorite stress-reduction techniques are running and cross-stitching. Both are pretty meditative and can clear my head. Productively stabbing fabric 1,000-plus times to make art takes a lot of concentration and patience, and there’s something really satisfying about seeing the end result.

Q: If you hadn’t been admitted to graduate school, what do you think you would be doing right now?

LM: If I couldn’t be in the biological sciences, I would probably be a meteorologist. We already joke I’m the lab weather person, so it feels like a natural extension!

Interested in Van Andel Institute Graduate School? Learn more at vaigs.vai.org and read previous student spotlights here.

The post Graduate student spotlight: Investigating a cancer linked to a rare genetic disorder appeared first on VAI.

New experimental blood test determines which pancreatic cancers will respond to treatment

$
0
0

GRAND RAPIDS, Mich. (Oct. 22, 2020) — Scientists have developed a simple, experimental blood test that distinguishes pancreatic cancers that respond to treatment from those that do not. This critical distinction could one day guide therapeutic decisions and spare patients with resistant cancers from undergoing unnecessary treatments with challenging side effects.

The findings were published today in Clinical Cancer Research, a journal of the American Association for Cancer Research.

Brian Haab, Ph.D.

“Knowing which type of pancreatic cancer a person has is critical to implementing the right treatment strategy for each patient,” said Brian Haab, Ph.D., a professor at Van Andel Institute and corresponding author of the study. “We hope that our new test, which detects a marker produced by cancer cells of one subtype and not the other, will one day soon be a powerful tool to help physicians and patients make the best decisions possible.”

Pancreatic cancers are among the most challenging malignancies to treat, due in part to their ability to evade detection until they have advanced and spread. Physicians currently have no reliable way to determine whether a patient has a subtype that will respond to existing chemotherapies versus a subtype that is resistant to treatment. The result often is a blanket treatment approach that works in only some but can have side effects in all.

The test detects and measures the levels of a sugar called sTRA, which is produced by a certain subtype of pancreatic cancer and escapes into the blood stream. Pancreatic cancers that produce sTRA tend to not respond to chemotherapy.

The new sTRA test evolved from an earlier test announced in January 2019. In that study, also published in Clinical Cancer Research, Haab and his colleagues described an experimental blood test that combined an existing diagnostic that detected a sugar called CA19-9 with a new test that detected sTRA. The combination approach detected nearly 70% of pancreatic cancers with a less than 5% false-positive rate — roughly 30% more than the CA19-9 alone. Both the 2019 combination test and the new sTRA test are slated to undergo additional clinical validation.

“The 2019 combination test tells us whether there is cancer and the new sTRA test helps us determine what kind of pancreatic cancer, which then could allow physicians to better narrow down the appropriate treatment plan,” Haab said. “When used in sequence, we believe the combination test and the new sTRA test could help catch and identify pancreatic cancer more quickly and definitively.”

Authors include ChongFeng Gao, Ph.D., Luke Wisniewski, Ying Liu, Ph.D., Ben Staal, Ian Beddows, Ph.D., Johnathan Hall and Daniel Barnett, Ph.D., of VAI; Dennis Plenker, Ph.D., Mirna Kheir Gouda and David A. Tuveson, M.D., Ph.D., of Cold Spring Harbor Laboratory; Mohammed Aldakkak, M.D., Douglas Evans, M.D., FACS, and Susan Tsai, M.D., MHS, of Medical College of Wisconsin; Peter Allen, M.D., of Duke University School of Medicine; Richard Drake, Ph.D., of Medical University of South Carolina; Amer Zureikat, M.D., Aatur Singhi, M.D., Ph.D., and Randall E. Brand, M.D., of University of Pittsburgh Medical Center; and Ying Huang, Ph.D., of Fred Hutchinson Cancer Research Center. VAI’s Optical Imaging Core, Bioinformatics and Biostatistics Core, Genomics Core and Pathology and Biorepository Core also supported this work.

Research reported in this publication was supported by Van Andel Institute; the National Cancer Institute of the National Institutes of Health under award no. U01CA152653 (Haab and Brand) and award no. U01CA226158 (Haab); the Lustgarten Foundation (Tuveson); and the German Research Foundation (Plenker). The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

###

ABOUT VAN ANDEL INSTITUTE
Van Andel Institute (VAI) is committed to improving the health and enhancing the lives of current and future generations through cutting edge biomedical research and innovative educational offerings. Established in Grand Rapids, Michigan, in 1996 by the Van Andel family, VAI is now home to more than 400 scientists, educators and support staff, who work with a growing number of national and international collaborators to foster discovery. The Institute’s scientists study the origins of cancer, Parkinson’s and other diseases and translate their findings into breakthrough prevention and treatment strategies. Our educators develop inquiry-based approaches for K-12 education to help students and teachers prepare the next generation of problem-solvers, while our Graduate School offers a rigorous, research-intensive Ph.D. program in molecular and cellular biology. Learn more at vai.org.

Media Contact
Beth Hinshaw Hall
Van Andel Institute
Beth.HinshawHall@vai.org
616-822-2064

The post New experimental blood test determines which pancreatic cancers will respond to treatment appeared first on VAI.

A look back at 2020

$
0
0

The past year has challenged us in new and unexpected ways. Yet through it all, we have been inspired by the strength and resilience of our community. We are deeply grateful to be part of West Michigan.

As with so many organizations and individuals, the pandemic upended many well-laid plans here at Van Andel Institute. But it could not stop the progress of scientists and the passion of educators, who have made vital advances despite unprecedented difficulties. 

Making strides in cancer

Rather than one disease, cancer refers to a group of more than 100 different diseases, all with one thing in common — uncontrolled cell growth. It represents a complex problem that requires an equally sophisticated approach in order to find new treatments and, hopefully, cures. That’s why VAI scientists tackle cancer from all angles, from studying its molecular underpinnings to investigating its roots in the genetic code to supporting clinical trials designed to find new treatments. Here are some highlights from their efforts in 2020:

  • Brian Haab and collaborators are developing a simple, experimental blood test that distinguishes pancreatic cancers that respond to treatment from those that do not. This critical distinction could one day guide therapeutic decisions and spare patients with resistant cancers from undergoing unnecessary treatments with challenging side effects. The experimental test is slated to undergo additional clinical validation. [1]
    Read more
  • VAI’s Biorepository was awarded a $2.7 million, two-year subcontract from the Frederick National Laboratory for Cancer Research currently operated by Leidos Biomedical Research, Inc. on behalf of the National Cancer Institute to serve as the biorepository for the Cancer Moonshot Biobank study, a national initiative to transform cancer treatment and prevention through accelerated research. [2]
    Read more
  • The American Association for Cancer Research awarded Van Andel Institute Professor Peter W. Laird, Ph.D., Director’s Scholar Stephen B. Baylin, M.D., FAACR, and Associate Professor Hui Shen, Ph.D., 2020 AACR Team Science Awards for their pivotal roles in the establishment and success of The Cancer Genome Atlas (TCGA), a landmark project that revolutionized our understanding of cancer and that is hailed as an exemplar of scientific collaboration.
    Read more

 Pursuing progress in Parkinson’s and other neurological disorders

Over the next decade, the incidences of Parkinson’s and Alzheimer’s are expected to increase significantly, reports the Parkinson’s Foundation and Alzheimer’s Association, respectively. Currently, there are no effective ways to slow or stop progression of either disease. At VAI, scientists are delving into the mechanisms that give rise to these diseases in hopes of providing a foundation for new treatments that halt progression and give people more years with fewer symptoms. Highlights from the past year include:

  • Can COVID-19 infection increase the risk of developing Parkinson’s disease? That’s the question posed by a commentary published in the journal Trends in Neurosciences, which explores three known case studies of people developing Parkinson’s-like symptoms in the weeks following infection with SARS-CoV-2, the virus that causes COVID-19. While rare, these cases provide important insights into potential long-term implications of infections. The commentary was co-authored by Dr. Patrik Brundin of VAI, Dr. Avindra Nath of the National Institute of Neurological Disorders and Stroke of the National Institutes of Health, and Dr. David Beckham of University of Colorado. [3]
    Read more
  • A collaborative team between the University of Minnesota Medical School and VAI was awarded $6.2 million for a study that seeks to define the molecular linkages between aging and Parkinson’s disease — an approach for new treatment targets not yet explored by many researchers. The three-year grant comes from the Aligning Science Across Parkinson’s initiative, an international collaborative research effort partnering with The Michael J. Fox Foundation for Parkinson’s Research to implement its funding. The study will combine four labs — Dr. Michael Lee and Dr. Laura Niedernhofer from University of Minnesota, and Dr. Darren Moore and Dr. José Brás from VAI.
    Read more
  • Rita Guerreiro and Dr. José Brás were awarded a $3.7 million grant from the National Institute on Aging to study the genetic predisposition to Alzheimer’s in the Portuguese population. The study will be the first of its kind in the country. [4]
    Read more
  • Switching off a molecular “master regulator” may protect the brain from inflammatory damage and neurodegeneration in Parkinson’s disease, according to a study led by Dr. Viviane Labrie. The study is the first of its kind and points to an entirely new avenue for developing therapies that preserve vulnerable brain cells in Parkinson’s disease. [5]
    Read more
  • A team led by Dr. Viviane Labrie may have solved one of the most puzzling and persistent mysteries in neuroscience: why some people are “right-brained” while others are “left-brained.” The answer lies in how certain genes on each side of the brain are switched “on” and “off” through a process called epigenetic regulation. The findings may explain why Parkinson’s disease and other neurological disorders frequently affect one side of the body first, a revelation that has far-reaching implications for development of potential future treatments. [6]
    Read more

Understanding how metabolism fuels health and disease

Metabolism provides fuel for each one of our bodies’ functions, from powering individual cells to ensuring we have the energy to complete our daily tasks. It is intricately linked to health and disease. By investigating how metabolism works and its links to other critical systems, such as the immune system, VAI scientists are charting a path toward a healthier tomorrow. Highlights from 2020 include:

  • A newly identified biomarker could help scientists pinpoint which cancers are vulnerable to treatment with biguanides, a common class of medications used to control blood sugar in Type 2 diabetes, reports a study led by VAI’s Dr. Russell Jones. Biguanides, particularly a medication called metformin, have long been of interest to cancer researchers because of their ability to target cellular metabolism, which fuels the growth and spread of malignant cells. [7]
    Read more
  • The immune system’s ability to marshal specialized cells to fight off infection relies in part on tiny molecules called microRNAs, which act as a release for the “brakes” that keep cells dormant until needed, according to a research team led by VAI’s Dr. Connie Krawczyk. Their findings revealed new insights into the nuts and bolts of immune function and add to a growing body of knowledge that could one day be leveraged to optimize vaccines or immunotherapies for a number of diseases. [8]
    Read more
  • Significantly reducing dietary levels of the amino acid methionine could slow onset and progression of inflammatory and autoimmune disorders such as multiple sclerosis in high-risk individuals. The findings — an important step toward potential future treatments — must be verified in humans first, said Dr. Russell Jones, who led the study. [9]
    Read more

Zooming in on life’s building blocks

VAI is home to one of the most powerful types of microscopes in the world, which allows our scientists to study life’s tiniest building blocks in exquisite detail. In fact, they are the same types of microscopes used by scientists in the U.S. and abroad to give us detailed glimpses of the virus that causes COVID-19. At VAI, our scientists use this groundbreaking technology, called cryo-electron microscopy, to visualize proteins and other important molecular structures that play important roles in health and disease. Understanding what these molecules look like gives us vital insights into how they work and how they may be targeted by new medications to treat a host of problems, such as cancer, neurological disorders and many others. Here are some highlights from the past year:

  • PAC, a class of molecular “gates” that maintain pH balance in cells, which helps keep cells alive and helps prevent stroke and other brain injuries. [10]
    Read more
  • PANX1, a molecular pathway that plays critical roles in human development, blood pressure regulation, inflammation and cell death. [11]
    Read more
  • EMC, a molecular “machine” that is responsible for installing signaling proteins into cellular membranes. The findings lay the foundation for potential future therapies for diseases like cancer, Alzheimer’s and cystic fibrosis. [12]
    Read more

Education

Student and teacher programs offered by Van Andel Institute for Education transitioned to a virtual format this year, including the implementation of webinars to support teachers as they transitioned to distance learning amid the pandemic. Education also rolled out a free project-based learning unit on how to prevent the spread of the coronavirus as well as mini-lessons on timely topics from respectful debate to the winter solstice.
Read more

Graduate School

Van Andel Institute Graduate School student Maggie Chassé earned a prestigious Predoctoral to Postdoctoral Fellow Transition Award from the National Cancer Institute of the National Institutes of Health. The award, also known as the F99/K00, provides up to two years of financial support for Ph.D. candidates to complete their dissertation research, and up to four years of support for postdoctoral training. 2020 was the second consecutive year a Graduate School student received an F99/K00 award.
Read more

Earlier in the year, the Graduate School welcomed two new team members to the organization: John Vasquez, Ph.D., as director of assessment and professional development, and Allison Roman as director of student support services. These hires, coupled with the construction of a new nearby building to accommodate larger incoming cohorts, signal the growing strength of the organization.
Read more

A number of Graduate School students were asked by Spectrum Health to participate in a COVID-19 literature review early in the pandemic. Many students remained engaged throughout the year in the process, which included reading material and providing feedback on which scientific articles may benefit the physicians treating patients.
Read more

Memorials

Dr. Viviane Labrie passed away in a tragic vehicle accident Aug. 21, 2020. Her ability to look at the world through different lenses allowed her to see old problems in new ways, and ultimately revealed groundbreaking insights with the potential to change lives. Dr. Labrie quickly established herself as a globally recognized leader in her field, holding her first faculty position in Toronto before joining VAI in 2016 as an assistant professor. She rapidly advanced, earning an early promotion to associate professor in 2019. Dr. Labrie earned numerous scientific awards and honors, including highly competitive grants from the National Institutes of Health and Department of Defense. Although early in her career, she already had made groundbreaking discoveries that are transforming the understanding of Parkinson’s and Alzheimer’s diseases, including the revelation that the appendix may be a starting point for Parkinson’s. Her findings led to exciting new avenues of discovery for potential treatments for these diseases, and shed light on the underpinnings of many other conditions, including bipolar disorder, schizophrenia and lactose intolerance. Dr. Labrie made an indelible mark on science, her colleagues and those she mentored, and her impact will be felt for years to come.

Dr. Luis Tomatis, a driving force in bringing VAI to life, passed away Sept. 29, 2020. Originally from Argentina, Dr. Tomatis was an energetic advocate for Grand Rapids. He was influential in creating and maintaining the environment that enabled the Institute to thrive and the Medical Mile to sprout up around us. Dr. Tomatis helped recruit top-tier scientific talent to establish VAI’s first Board of Scientific Advisors and appoint Nobel Laureate Dr. Michael Brown as the board’s first chairman. After serving as VAI’s founding president from 1995–2001, he went on to become the director of medical affairs for the Richard M. DeVos Family. A cardiothoracic surgeon, Dr. Tomatis was a former chief of cardiovascular surgery at Spectrum Health and MSU professor of cardiac surgery. He won numerous awards, volunteered for organizations like the American Heart Association and the Grand Rapids Symphony, and was responsible for arranging for invaluable medical equipment to be sent to his homeland of Argentina.

Dr. Gordon Van Wylen passed away Nov. 5, 2020, at the age of 100. As one of VAI’s original trustees, Dr. Van Wylen was instrumental in establishing the early programs of the Education Institute and created the groundwork for the success of VAI. We continue to see his vision in action through the programs we are developing and implementing today, some 25 years later. A man of immense scientific knowledge, education, experience and integrity, Dr. Van Wylen was an accomplished educator and administrator. He served as dean of engineering at University of Michigan, as Hope College president for 15 years, and as founding trustee and inaugural director of Van Andel Institute for Education. Dr. Van Wylen generously devoted his time and talent to numerous other organizations throughout his life. He was widely regarded as a thoughtful, visionary, kind and respectful leader.

Funding

[1] Research reported in this publication was supported by Van Andel Institute; the National Cancer Institute of the National Institutes of Health under award no. U01CA152653 (Haab and Brand) and award no. U01CA226158 (Haab); the Lustgarten Foundation (Tuveson); and the German Research Foundation (Plenker). The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

[2] The project has been funded in whole or in part with federal funds from the National Cancer Institute of the National Institutes of Health under contract no. HHSN261201500003I, Task Order HHSN26100042 through Leidos Biomedical Research, Inc. under subcontract no. 20X062Q. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the U.S. government.

[3] Research reported in this publication was supported by Van Andel Institute; the Farmer Family Foundation (Brundin); Division of Intramural Research, National Institute of Neurological Disorders and Stroke of the National Institutes of Health (Nath); and a VA Merit Award (Beckham). The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

[4] Research reported in this publication is supported by the National Institute on Aging of the National Institutes of Health under award no. R01AG067426. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

[5] The U.S. Army Medical Research Acquisition Activity, 820 Chandler Street, Fort Detrick MD 21702-5014 is the awarding and administering acquisition office. This work was supported by the Office of the Assistant Secretary of Defense for Health Affairs, through the Congressionally Directed Medical Research Programs’ Parkinson’s Research Program under Award No. W81XWH1810512 (Labrie). Opinions, interpretations, conclusions and recommendations are those of the author and are not necessarily endorsed by the Department of Defense. In conducting research using animals, the investigators adhere to the laws of the United States and regulations of the Department of Agriculture. Animal research at VAI is conducted in accordance with the National Institutes of Health’s Office of Laboratory Animal Welfare Public Health Service Policy on Humane Care and Use of Laboratory Animals. VAI also is accredited by AAALAC International. This work also was supported by a VAI Innovation Award (Labrie). Labrie also has awards from the National Institute of Neurological Disorders and Stroke of the National Institutes of Health and Michigan State University through the Gibby & Friends vs. Parky Parkinson’s Disease Research Award.

[6] This work was supported by Van Andel Institute. Labrie is supported by the U.S. Army Medical Research Materiel Command through the Parkinson’s Research Program Investigator-Initiated Research Award under award no. W81XWH1810512. Opinions, interpretations, conclusions and recommendations are those of the author and are not necessarily endorsed by the U.S. Army. Labrie also is supported by the National Institute of Neurological Disorders and Stroke of the National Institutes of Health under Award Number R21NS112614 and by Michigan State University through the Gibby & Friends vs. Parky Parkinson’s Disease Research Award. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or other granting organizations.

[7] Research reported in this publication was supported by the Canadian Institutes of Health Research under grants MOP-142259 (Jones) and MOP-123352 (Duchaine); The Medical Research Council under grant MC_UU_0015/2 (Hirst); and funding from ImmunoMet Therapeutics. The Goodman Cancer Research Center Metabolomics Core Facility is supported by grants from the Canadian Foundation for Innovation, Canadian Institutes of Health Research and Terry Fox Research Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

[8] Research reported in this publication was supported by the National Sciences and Engineering Research Council (NSERC) under grant nos. RGPIN/2018-06257 and RGPIN/419537-2012 (Krawczyk). Brendan Cordeiro was supported by the McGill Integrated Cancer Research Training Program, the Fonds de la Rescherche du Quebec-Santé and the Canadian Institutes of Health Research. The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations. 

[9] Research reported in this publication was supported by the Multiple Sclerosis Society of Canada (Larochelle) and the Canadian Institutes of Health Research under award nos. MOP-399061 (Witcher) and MOP-142259 (Jones). The content is solely the responsibility of the authors and does not necessarily represent the official views of any funding organization.

[10] Research reported in this publication was supported by Van Andel Institute; McKnight Scholar Awards in Neuroscience (Du, Qiu), Klingenstein-Simons Scholar Awards (Du, Qiu); Sloan Research Fellowships (Du, Qiu); the National Institute of General Medical Sciences of the National Institutes of Health under award no. R35GM124824 (Qiu); the National Institute of Neurological Disorders and Stroke of the National Institutes of Health under award no. R01NS118014 (Qiu), R01NS112363 (Lü) and R01NS111031 (Du); the National Heart, Lung and Blood Institute of the National Institutes of Health under award no. R56HL144929 (Lü); a Pew Scholar in Biomedical Sciences award (Du); and the American Heart Association under award no. 20POST35120556 (Ruan) and 18PRE34060025 (Osei-Owusu). The content is solely the responsibility of the authors and does not necessarily represent the official views of the granting organizations.

[11] Research reported in this publication was supported by Van Andel Institute.

Lü is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under grant no. R56HL144929. Du is supported by the National Institute of Neurological Disorders and Stroke of the National Institutes of Health under grant no. R01NS111031, a McKnight Scholar Award, a Klingenstein-Simons Scholar Award and a Sloan Research Fellowship in neuroscience. Ruan is supported by an American Heart Association postdoctoral fellowship for his study on the PANX1 channel. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or other granting organizations.

[12] Research reported in this publication was supported by Van Andel Institute and the National Cancer Institute of the National Institutes of Health under award no. CA231466 (Li). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

The post A look back at 2020 appeared first on VAI.

What you need to know about colorectal cancers

$
0
0

In 2020, colorectal cancers accounted for 8%, or 147,950, of new cancer cases, making them the fourth most common type of new cancers diagnosed in the U.S. (excluding skin cancers). 1Colon and rectal cancers, often referred together as “colorectal cancer,” share some important similarities — both affect parts of the large intestine, both frequently present as adenocarcinomas (a type of cancer that begins in cells that produce fluids such as mucus), and both start as growths called polyps.2 Colorectal cancers are the second leading cause of cancer-related death in the U.S. after lung cancer.1

Colorectal cancer cases are on the rise in young people.

The majority of colorectal cancers are diagnosed in people older than age 50, the age many experts recommend people with average risk begin screening.3

However, in 2017, American Cancer Society (ACS) researchers reported a disturbing increase in the number of cases diagnosed in younger people — namely, that people born in 1990 have double the risk of developing colon cancer and quadruple the risk of developing rectal cancer than people born in 1950. As a result, ACS lowered their recommended age for screening to 45 for people with average risk.4

While the rate of new cases for these diseases has been dropping overall — about 2.7% annually over the past decade, according to the National Cancer Institute — the ACS study revealed that this decline is largely fueled by older people. When they broke down the data, they found that incidence rates (rates of new cases) have actually been increasing to the tune of 1% to 2% each year for colon cancer in people ages 20 to 39 and 3% per year for rectal cancer in adults ages 20 to 29.4

As of 2020, 12% (or 18,000 cases) of colorectal cancer were estimated to be diagnosed in people younger than age 50.5

It’s not entirely clear why this increase is occurring, but it’s likely linked in part to obesity, poor diet and lack of exercise. To lower your risk, experts recommend4:

  • Regularly exercising
  • Eating a balanced diet (more whole grains, fruits and vegetables, and less red meat and processed meats)
  • Avoiding smoking and limiting alcohol consumption

In many cases, symptoms aren’t apparent in the earliest stages of the disease.
People younger than age 55 are 58% more likely to be diagnosed with late-stage cancer, in part because “cancer is typically not on the radar of young adults and their providers,” the authors of the ACS study said at the time.4

Early detection is critical and may prevent the disease entirely if precancerous polyps are found and removed. People should be vigilant about symptoms and discuss any concerns with their physician. Signs include:4

  • A prolonged change in bowel habits
  • Cramping or abdominal pain
  • Rectal bleeding, blood in the stool or dark stools
  • A feeling that you have to go that is not alleviated by a bowel movement
  • Weakness, fatigue or unintended weight loss

Experts recommend talking to your doctor if you have a family history of colorectal cancers. Due to a higher incidence of developing colorectal cancers, many experts also recommend African Americans begin screening early at age 45.6

Read more about colorectal cancer research at Van Andel Institute at the links below:

 

 

Sources

1 National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Common Cancer Sites. Accessed February 9, 2021. seer.cancer.gov/statfacts/html/common.html

2 National Cancer Institute. Colorectal Cancer — Patient Version. Accessed February 9, 2021.  www.cancer.gov/types/colorectal

3 Centers for Disease Control and Prevention. Colorectal Cancer Screening Tests. Updated: February 10, 2020. Retrieved February 9, 2021. www.cdc.gov/cancer/colorectal/basic_info/screening/tests.htm

4 Study finds sharp rise in colon cancer and rectal cancer rates among young adults. American Cancer Society. February 28, 2017. Accessed February 9, 2021. www.cancer.org/latest-news/study-finds-sharp-rise-in-colon-cancer-and-rectal-cancer-rates-among-young-adults.html

5 American Cancer Society. Colorectal cancer rates rise in younger adults. March, 5, 2020. Accessed February 9, 2021. www.cancer.org/latest-news/colorectal-cancer-rates-rise-in-younger-adults.html

6 American Cancer Society. Colorectal cancer rates higher in African Americans, rising in younger people. September 3, 2020. Accessed February 9, 2021.

The post What you need to know about colorectal cancers appeared first on VAI.

Viewing all 60 articles
Browse latest View live




Latest Images