Sharpening the edges of cancer chemotherapy
Nicole Davis | Whitehead Institute
July 11, 2018

Cambridge, MA — Tackling unsolved problems is a cornerstone of scientific research, propelled by the power and promise of new technologies. Indeed, one of the shiniest tools in the biomedical toolkit these days is the genome editing system known as CRISPR/Cas9. Whitehead Institute Member David Sabatini and his colleagues pioneered the use of this tool as a foundation for large-scale genetic screens in human cells, turning up a treasure trove of new insights into cellular metabolism, in both normal cells and cancer cells.

When Naama Kanarek, a postdoc in Sabatini’s laboratory, pondered how to apply these state-of-the-art CRISPR/Cas9 screens to her own research, her thoughts turned to a classic cancer chemotherapy drug, methotrexate, which has been in clinical use for nearly seven decades. Often used to treat a form of pediatric leukemia, known as acute lymphoblastic leukemia (ALL), the drug, when deployed as part of a multifaceted treatment plan, can be highly effective. But its power comes at a cost. Because methotrexate can damage not only cancer cells but also healthy tissues, it must be administered with great care. For children who receive high doses of the drug, a mainstay of ALL treatment, that can mean several days spent in the hospital with rigorous clinical monitoring.

In other forms of cancer, methotrexate’s efficacy is more uncertain. For example, in pediatric osteosarcoma, only 65 percent of patients respond. Unfortunately, there is currently no way for doctors to pinpoint who will and who will not.

“From a scientific standpoint, methotrexate is quite special because it was the first metabolic drug to be developed, but much of its biology remains to be discovered — particularly what drives these different responses in patients,” Kanarek says. “So, this is really one of these old, classic questions that has been lingering in the field for some time. We thought we could learn something new.”

And they did. In the July 11 online issue of the journal Nature, Kanarek, Sabatini, and their colleagues report the findings of a CRISPR/Cas9 screen for factors involved in methotrexate sensitivity. The team’s work yielded a surprising set of discoveries that point to the breakdown of histidine — one of several amino acids used by the body to construct proteins — as a critical gatekeeper of cancer cells’ vulnerability to methotrexate. The researchers’ findings not only help illuminate the biology of a well-known cancer chemotherapy, but also suggest a simple dietary supplement that could help broaden its therapeutic window and reduce its toxicity.

 “This study is an example of the power of modern genomic tools to shine a bright light on longstanding questions in human biology,” says senior author David Sabatini, a Member of Whitehead Institute, a professor of biology at Massachusetts Institute of Technology and investigator with the Howard Hughes Medical Institute (HHMI). “While cancer chemotherapies can be quite effective, their biological effects are often poorly understood. By laying bare their biology, we may be able to devise ways to utilize them more wisely.”

ATTACK THE CANCER, NOT THE PATIENT

The history of methotrexate stretches back to the 1940s, a time when strikingly little was known about the origins of cancer much less how best to treat it. The birth of methotrexate as a chemotherapeutic agent was sparked by the astute observations of Sidney Farber, a pediatric pathologist at Boston Children’s Hospital who cared for children with a variety of maladies, including ALL. In the course of caring for patients with ALL, Farber recognized that cancer cells depended on the nutrient folic acid for their own proliferation. That gave him the idea of using folate antagonists to treat ALL. Methotrexate was developed in 1949 precisely for this purpose and was subsequently shown to induce remission in children with ALL. Fast forward to today, and the drug has evolved into a significant tool in oncologists’ toolkit.

“Methotrexate is a major part of the backbone of chemotherapy treatment across many human cancers,” says Loren Walensky, a pediatric hematologist/oncologist at the Dana-Farber Cancer Institute who is not a study co-author but served as an early adviser on the project and will also play a deeper role in planning future follow-up studies. “It is also used outside of the cancer field for the treatment of several autoimmune diseases.”

He added, “But as with all chemotherapy, the critical issue is how to best use it to inflict maximal damage on the cancer without irreparably harming the patient.”

Kanarek explains how new genetic tools are allowing insights into the sensitivity of cancer cells to methotrexate.

The basic mechanics of methotrexate are fairly well known. The drug inhibits dihydrofolate reductase (DHFR), an enzyme that generates the functional form of folate, known as tetrahydrofolate (THF). THF is essential for preparing the raw materials needed to make nucleic acids, such as DNA, which carries cells’ genetic information, and RNA, a close chemical relative involved in making proteins. “Proliferating cells must duplicate their DNA, so they need a lot of THF,” Kanarek explains. “But even cells that are not dividing need to make RNA, and that requires THF, too.”

The results of Kanarek’s CRISPR/Cas9 screen now bring greater clarity to this molecular picture. She and her colleagues uncovered another enzyme, called FTCD, which is involved in the breakdown of histidine. Interestingly, FTCD also requires THF for its function — though not nearly as much as the main target of methotrexate, DHFR. Despite the differential demands of the two enzymes, they both draw from the same, shared pool of THF.

“Under normal conditions, this pool is sufficiently full, so there is no competition for resources, even in rapidly dividing cells,” Kanarek says.

But when the amount of THF becomes limiting — as it does in cells that are treated with methotrexate — the story is quite different, the Whitehead Institute team discovered. In that case, the activity of FTCD poses serious problems, because there isn’t enough THF in the pool to support both cell proliferation and histidine breakdown. When that happens, the cells die.

That got Kanarek thinking more about histidine: Could the nutrient provide a way to tinker with FTCD activity and, by virtue of the cancer cells’ own metabolism, make them more vulnerable to methotrexate?

To explore this question, the researchers used mouse models of leukemia, engineered by transplanting human leukemia cells under the skin of immunocompromised mice. A subset of the mice received injections of methotrexate together with histidine. This one-two punch, Kanarek hypothesized, should ramp up the function of FTCD and more rapidly drain the THF pool, thereby making the cells more sensitive to the cancer-killing effects of methotrexate.

That is precisely what the team observed. Notably, these experiments involved lower than normal doses of methotrexate, suggesting the cells had indeed been made more sensitive to the cancer drug. Moreover, the studies included a human leukemia cell line, called SEM, which harbors a specific genetic mutation that is associated with a particularly poor prognosis in patients — further underscoring the power of the histidine degradation pathway to weaken cells’ defenses.

Now, Kanarek and her colleagues are working to extend these initial findings with additional preclinical studies and, together with Walensky, determine how to best evaluate the potential benefits of histidine supplementation in cancer patients. Their ultimate goal: to pursue clinical trials that will assess histidine’s ability to improve the effectiveness of methotrexate in humans.

In addition to making cancer cells more vulnerable to methotrexate, the Whitehead Institute team’s research also holds promise for another therapeutic challenge: identifying which patients will or will not respond to the drug.

Two other enzymes cooperate with FTCD in breaking down histidine. The levels of one of the enzymes, known as HAL, appears to correlate with cells’ sensitivity to methotrexate: That is, cancer cells with high levels of HAL tend to be more sensitive to the drug. More work is needed to determine whether this correlation extends to a broader swath of patient samples and if it has predictive value in the clinic. Nevertheless, Kanarek and her colleagues are already beginning work on this front. Together with Abner Louissaint, Jr., a hematopathologist at Massachusetts General Hospital who also served as an early adviser on the Nature study, the Whitehead Institute team will launch a second clinical study to examine whether HAL levels can predict methotrexate response in patients with lymphoma.

“Being able to understand who is going to respond to methotrexate and who is not, and how to achieve a therapeutic benefit while mitigating the drug’s potential side effects, could have a profound impact on patient care,” Walensky says. “The insights from this study bring an entirely new dimension to our understanding of a decades-old and critically important cancer medicine. And as a physician and a scientist, that’s truly exciting.”

Written by Nicole Davis

* * *

David Sabatini’s primary affiliation is with Whitehead Institute for Biomedical Research, where his laboratory is located and all his research is conducted. He is also a Howard Hughes Medical Institute investigator and a professor of biology at Massachusetts Institute of Technology.

* * *

Full citation:

“Histidine catabolism is a major determinant of methotrexate sensitivity”

Nature, online on July 11, 2018.

Naama Kanarek (1,2,3,4), Heather R. Keys (1), Jason R. Cantor (1,2,3,4), Caroline A. Lewis (1), Sze Ham Chan (1), Tenzin Kunchok (1), Monther Abu-Remaileh (1,2,3,4), Elizaveta Freinkman (1), Lawrence D. Schweitzer (4), and David M. Sabatini (1,2,3,4).

  1. Whitehead Institute for Biomedical Research and Massachusetts Institute of Technology, Department of Biology, 455 main Street, Cambridge, Massachusetts 02142, USA
  2. Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
  3. Koch Institute for Integrative Cancer Research and Massachusetts Institute of Technology, Department of Biology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
  4. Broad Institute of Harvard and Massachusetts Institute of Technology, 415 main Street, Cambridge, Massachusetts 02142, USA
Restricting a key cellular nutrient could slow tumor growth

Researchers identify the amino acid aspartate as a metabolic limitation in certain cancers.

Raleigh McElvery | Department of Biology
June 29, 2018

Remove tumor cells from a living organism and place them in a dish, and they will multiply even faster than before. The mystery of why this is has long stumped cancer researchers, though many have simply focused on the mutations and chains of molecular reactions that could prompt such a disparity. Now, a group of MIT researchers suggests that the growth limitations in live organisms may stem from a different source: the cell’s environment. More specifically, they found that the amino acid aspartate serves as a key nutrient needed for the “proliferation” or rapid duplication of cancer cells when oxygen is not freely available.

The biologists took cancer cells from various tissue types and engineered them to convert another, more abundant substrate into aspartate using the gene encoding an enzyme from guinea pigs. This had no effect on the cells sitting in a dish, but the same cells implanted into mice engendered tumors that grew faster than ever before. The researchers had increased the cells’ aspartate supply, and in doing so successfully sped up proliferation in a living entity.

“There hasn’t been a lot of thought into what slows tumor growth in terms of the cellular environment, including the sort of food cancer cells need,” says Matthew Vander Heiden, associate professor of biology, associate director of the Koch Institute for Integrative Cancer Research, and senior author of the study. “For instance, if you’re trying to get to a given destination and I want to slow you down, my best bet is to set up a roadblock at a place on your route where you’d experience a slow-down anyways, like a long traffic light. That’s essentially what we’re interested in here — understanding what nutrients the cell is already lacking that put the brakes on proliferation, and then further limiting those nutrients to inhibit growth even more.”

Lucas Sullivan, a postdoc in Vander Heiden’s lab, is the lead author of the study, which appeared in Nature Cell Biology on June 25.

Building the case for aspartate

Isolating a single factor that could impact tumor growth within an organism is tricky business. One potential candidate came to Sullivan via a paper he co-authored with graduate student Dan Gui in 2015, which asked a somewhat controversial question: Why is it that cells need to consume oxygen through cellular respiration in order to proliferate?

It’s a rather counter-intuitive question, because some scientific literature suggests just the opposite: Cancer cells in an organism (“in vivo”) do not enjoy the same access to oxygen as they would in a dish, and therefore don’t depend on oxygen to produce enough energy to divide. Instead, they switch to a different process, fermentation, that doesn’t require oxygen. But Sullivan and Gui noted that cancer cells do rely on oxygen for another reason: to produce aspartate as a byproduct.

Aspartate, they soon confirmed, does, in fact, play a crucial role in controlling the rate of cancer cell proliferation. In another study one year later, Sullivan and Gui noted that the antidiabetic drug metformin, known to inhibit mitochondria, slowed tumor growth and decreased aspartate levels in cells in vivo. Since mitochondria are key to cellular respiration, Sullivan reasoned that blocking their function in an already oxygen-constrained environment (the tumor) might make cancer cells vulnerable to further suppression of respiration — and aspartate — explaining why metformin seems to have such a strong effect on tumor growth.

Despite being potentially required for certain amino acids and the synthesis of all four DNA nucleotides, aspartate is already hard to come by, even in oxygen-rich environments. It’s among the lowest concentration amino acids in our blood, and has no way to enter our cells unless a rare protein transporter is present. Precisely why aspartate import is so inefficient remains an evolutionary mystery; one possibility is that its scarcity serves as a “failsafe,” preventing cells from multiplying until they have all the resources to properly do so.

Regardless, the easiest way for cells to get aspartate is not to import it from outside, but rather to make it directly inside, breaking down another amino acid called asparagine to generate it. However, there are very few known mammals that have an enzyme capable of producing aspartate from asparagine — among them, the guinea pig.

Channeling the guinea pig

In the 1950s, a researcher named John Kidd made an accidental discovery. He injected cancer-ridden rats with sera from various animals — rabbits, horses, guinea pigs, and the like — and discovered that guinea pig serum alone shrunk the rats’ tumors. It wasn’t until years later that scientists learned it was an enzyme in the guinea pig blood called guinea pig asparaginase 1 (gpASNase1) that was responsible for this antitumorigenic effect. Today, we know about a host of simpler organisms with similar enzymes, including bacteria and zebrafish. In fact, bacterial asparaginase is approved as a medicine to treat acute lymphocytic leukemia.

Because guinea pigs are mammals and thus have similar metabolisms to our own, the MIT researchers decided to use gpASNase1 to increase aspartate levels in tumors in four different tumor types and ask whether the tumors would grow faster. This was the case for three of the four types: The colon cancer cells, osteosarcoma cells, and mouse pancreatic cancer cells divided more rapidly than before, but the human pancreatic cancer cells continued to proliferate at their normal pace.

“This is a relatively small sample, but you could take this to mean that not every cell in the body is as sensitive to loss of aspartate production as others,” Sullivan says. “Acquiring aspartate may be a metabolic limitation for only a subset of cancers, since aspartate can be produced via a number of different pathways, not just through asparagine conversion.”

When the researchers tried to slow tumor growth using the antidiabetic metformin, the cells expressing gpASNase1 remained unaffected — confirming Sullivan’s prior suspicion that metformin slows tumor growth specifically by impeding cellular respiration and suppressing aspartate production.

“Our initial finding connecting metformin and proliferation was very serendipitous,” he says, “but these most recent results are a clear proof of concept. They show that decreasing aspartate levels also decreases tumor growth, at least in some tumors. The next step is to determine if there are other ways to more intentionally target aspartate synthesis in certain tissues and improve our current therapeutic approaches.”

Although the efficacy of using metformin to treat cancer remains controversial, these findings indicate that one means to target tumors would be to prevent them from accessing or producing nutrients like aspartate to make new cells.

“Although there are many limitations to cancer cell proliferation, which metabolites become limiting for tumor growth has been poorly understood,” says Kivanc Birsoy, the Chapman-Perelman Assistant Professor at Rockefeller University. “This study identifies aspartate as one such limiting metabolite, and suggests that its availability could be targeted for anti-cancer therapies.”

Birsoy is a former postdoc in professor of biology David Sabatini’s lab, who authored a paper published in the same issue of Nature Cell Biology, identifying aspartate as a major growth limitation in oxygen-deprived tumors.

“These companion papers demonstrate that some tumors in vivo are really limited by the chemical processes that require oxygen to get the aspartate they need to grow, which can affect their sensitivity to drugs like metformin,” Vander Heiden says. “We’re beginning to realize that understanding which cancer patients will respond to which treatments may be determined by factors besides genetic mutations. To really get the full picture, we need to take into account where the tumor is located, its nutrient availability, and the environment in which it lives.”

The research was funded by an NIH Pathway to Independence Award, the American Cancer Society, Ludwig Center for Molecular Oncology Fund, the National Science Foundation, a National Institutes of Health Ruth Kirschstein Fellowship, Alex’s Lemonade Stand Undergraduate Research Fellowship, Damon Runyon Cancer Research Foundation, Howard Hughes Medical Institute Faculty Scholar Award, Stand Up to Cancer, Lustgarten Foundation, Ludwig Center at MIT, the National Institutes of Health, and the Koch Institute’s Center for Precision Cancer Medicine.

Fasting boosts stem cells’ regenerative capacity

A drug treatment that mimics fasting can also provide the same benefit, study finds.

Anne Trafton | MIT News Office
May 1, 2018

As people age, their intestinal stem cells begin to lose their ability to regenerate. These stem cells are the source for all new intestinal cells, so this decline can make it more difficult to recover from gastrointestinal infections or other conditions that affect the intestine.

This age-related loss of stem cell function can be reversed by a 24-hour fast, according to a new study from MIT biologists. The researchers found that fasting dramatically improves stem cells’ ability to regenerate, in both aged and young mice.

In fasting mice, cells begin breaking down fatty acids instead of glucose, a change that stimulates the stem cells to become more regenerative. The researchers found that they could also boost regeneration with a molecule that activates the same metabolic switch. Such an intervention could potentially help older people recovering from GI infections or cancer patients undergoing chemotherapy, the researchers say.

“Fasting has many effects in the intestine, which include boosting regeneration as well as potential uses in any type of ailment that impinges on the intestine, such as infections or cancers,” says Omer Yilmaz, an MIT assistant professor of biology, a member of the Koch Institute for Integrative Cancer Research, and one of the senior authors of the study. “Understanding how fasting improves overall health, including the role of adult stem cells in intestinal regeneration, in repair, and in aging, is a fundamental interest of my laboratory.”

David Sabatini, an MIT professor of biology and member of the Whitehead Institute for Biomedical Research and the Koch Institute, is also a senior author of the paper, which appears in the May 3 issue of Cell Stem Cell.

“This study provided evidence that fasting induces a metabolic switch in the intestinal stem cells, from utilizing carbohydrates to burning fat,” Sabatini says. “Interestingly, switching these cells to fatty acid oxidation enhanced their function significantly. Pharmacological targeting of this pathway may provide a therapeutic opportunity to improve tissue homeostasis in age-associated pathologies.”

The paper’s lead authors are Whitehead Institute postdoc Maria Mihaylova and Koch Institute postdoc Chia-Wei Cheng.

Boosting regeneration

For many decades, scientists have known that low caloric intake is linked with enhanced longevity in humans and other organisms. Yilmaz and his colleagues were interested in exploring how fasting exerts its effects at the molecular level, specifically in the intestine.

Intestinal stem cells are responsible for maintaining the lining of the intestine, which typically renews itself every five days. When an injury or infection occurs, stem cells are key to repairing any damage. As people age, the regenerative abilities of these intestinal stem cells decline, so it takes longer for the intestine to recover.

“Intestinal stem cells are the workhorses of the intestine that give rise to more stem cells and to all of the various differentiated cell types of the intestine. Notably, during aging, intestinal stem function declines, which impairs the ability of the intestine to repair itself after damage,” Yilmaz says. “In this line of investigation, we focused on understanding how a 24-hour fast enhances the function of young and old intestinal stem cells.”

After mice fasted for 24 hours, the researchers removed intestinal stem cells and grew them in a culture dish, allowing them to determine whether the cells can give rise to “mini-intestines” known as organoids.

The researchers found that stem cells from the fasting mice doubled their regenerative capacity.

“It was very obvious that fasting had this really immense effect on the ability of intestinal crypts to form more organoids, which is stem-cell-driven,” Mihaylova says. “This was something that we saw in both the young mice and the aged mice, and we really wanted to understand the molecular mechanisms driving this.”

Metabolic switch

Further studies, including sequencing the messenger RNA of stem cells from the mice that fasted, revealed that fasting induces cells to switch from their usual metabolism, which burns carbohydrates such as sugars, to metabolizing fatty acids. This switch occurs through the activation of transcription factors called PPARs, which turn on many genes that are involved in metabolizing fatty acids.

The researchers found that if they turned off this pathway, fasting could no longer boost regeneration. They now plan to study how this metabolic switch provokes stem cells to enhance their regenerative abilities.

They also found that they could reproduce the beneficial effects of fasting by treating mice with a molecule that mimics the effects of PPARs. “That was also very surprising,” Cheng says. “Just activating one metabolic pathway is sufficient to reverse certain age phenotypes.”

Jared Rutter, a professor of biochemistry at the University of Utah School of Medicine, described the findings as “interesting and important.”

“This paper shows that fasting causes a metabolic change in the stem cells that reside in this organ and thereby changes their behavior to promote more cell division. In a beautiful set of experiments, the authors subvert the system by causing those metabolic changes without fasting and see similar effects,” says Rutter, who was not involved in the research. “This work fits into a rapidly growing field that is demonstrating that nutrition and metabolism has profound effects on the behavior of cells and this can predispose for human disease.”

The findings suggest that drug treatment could stimulate regeneration without requiring patients to fast, which is difficult for most people. One group that could benefit from such treatment is cancer patients who are receiving chemotherapy, which often harms intestinal cells. It could also benefit older people who experience intestinal infections or other gastrointestinal disorders that can damage the lining of the intestine.

The researchers plan to explore the potential effectiveness of such treatments, and they also hope to study whether fasting affects regenerative abilities in stem cells in other types of tissue.

The research was funded by the National Institutes of Health, the V Foundation, a Sidney Kimmel Scholar Award, a Pew-Stewart Trust Scholar Award, the Kathy and Curt Marble Cancer Research Fund, the MIT Stem Cell Initiative through Fondation MIT, the Koch Institute Frontier Research Program through the Kathy and Curt Marble Cancer Research Fund, the American Federation of Aging Research, the Damon Runyon Cancer Research Foundation, the Robert Black Charitable Foundation, a Koch Institute Ludwig Postdoctoral Fellowship, a Glenn/AFAR Breakthroughs in Gerontology Award, and the Howard Hughes Medical Institute.

Study suggests perioperative NSAIDs may prevent early metastatic relapse in post-surgical breast cancer patients
Nicole Giese Rura | Whitehead Institute
April 11, 2018

Cambridge, MA – According to research conducted in mice by Whitehead Institute scientists, surgery in breast cancer patients, which while often curative, may trigger a systemic immunosuppressive response, allowing the outgrowth of dormant cancer cells at distant sites whose ability to generate tumors had previously been kept in check by the immune system. Taking a non-steroidal anti-inflammatory drug (NSAID) around the time of surgery may thwart such early metastatic relapse without impeding post-surgical wound healing.

The team’s work was published in the April 11 issue of the journal Science Translational Medicine.

“This represents the first causative evidence of surgery having this kind of systemic response,” says Jordan Krall, the first author of the paper and a former postdoctoral researcher in the lab of Whitehead Founding Member Robert Weinberg. “Surgery is essential for treating a lot of tumors, especially breast cancer. But there are some side effects of surgery, just as there are side effects to any treatment.  We’re starting to understand what appears to be one of those potential side effects, and this could lead to supportive treatment alongside of surgery that could mitigate some of those effects.”

Although the association between surgery and metastatic relapse has been documented, a causal line between the two has never been established, leading many to consider early metastatic relapse to be the natural disease progression in some patients. Previous studies of breast cancer patients have shown a marked peak in metastatic relapse 12-18 months following surgery. Although the underlying mechanism for such a spike has not been understood, a 2010 retrospective clinical trial conducted in Belgium provides a clue: Breast cancer patients taking a non-steroidal anti-inflammatory (NSAID) for pain following tumor resection had lower rates of this early type of metastatic relapse than patients taking opioids for post-surgical pain. Anti-inflammatory drugs also have previously been shown to directly inhibit tumor growth, but Krall and Weinberg thought that the NSAIDs’ effects in these studies may be independent of the mechanism responsible for the effects noted in the retrospective clinical trial.

To investigate the causes of early metastatic relapse after surgery, the team created a mouse model that seems to mirror the immunological detente keeping in check dormant, disseminated tumor cells in breast cancer patients. In this experimental model, the mice’s T cells stall the growth of tumors that are seeded by injected cancer cells. When mice harboring dormant cancer cells underwent simulated surgeries at sites distant from the tumor cells, tumor incidence and size dramatically increased. Analysis of the blood and tumors from wounded mice showed that wound healing increases levels of cells called inflammatory monocytes, which differentiate into tumor-associated macrophages.  Such macrophages, in turn, can act at distant sites to suppress the actions of T lymphocytes that previously succeeded in keeping the implanted tumors under control. Krall and Weinberg then tested the effects of the NSAID meloxicam (Mobic®), thinking that this anti-inflammatory drug might block the effects of immuno-suppressive effects of wound healing.  In fact, when mice received the NSAID after or at the time of surgery, the drug prevented a systemic inflammatory response created by the wound healing and the meloxicam-treated mice developed significantly smaller tumors than wounded, untreated mice; often these tumors completely disappeared. Notably, meloxicam did not impede the mice’s wound healing

Still, Weinberg cautions that scientists are just beginning to understand the connections between post-surgical wound healing, inflammation, and metastasis.

“This is an important first step in exploring the potential importance of this mechanism in oncology,” says Weinberg, who is also a professor of biology at Massachusetts Institute of Technology (MIT) and director of the MIT/Ludwig Center for Molecular Oncology.

This work was supported by the Advanced Medical Research Foundation, the Transcend Program (a partnership between the Koch Institute and Janssen Pharmaceuticals Inc.), the Breast Cancer Research Foundation, the Ludwig Center for Molecular Oncology at MIT, and the Samuel Waxman Cancer Research Foundation, the American Cancer Society, Hope Funds for Cancer Research, the Charles A. King Trust, the National Health and Medical Research Council of Australia (NHMRC APP1071853), the National Institutes of Health (NIH/NCI 1K99CA201574-01A1), the American Cancer Society Ellison Foundation (PF-15-131-01-CSM), and the U.S. Department of Defense (W81XWH-10-1-0647).

* * *
Robert Weinberg’s primary affiliation is with Whitehead Institute for Biomedical Research, where his laboratory is located and all his research is conducted. He is also a professor of biology at Massachusetts Institute of Technology and director of the MIT/Ludwig Center for Molecular Oncology.
* * *
Full Citation:
“The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy”
Science Translational Medicine, April 11, 2018.
 Jordan A. Krall (1), Ferenc Reinhardt (1), Oblaise A. Mercury (1), Diwakar R. Pattabiraman (1), Mary W. Brooks (1), Michael Dougan (1,2), Arthur W. Lambert (1), Brian Bierie (1), Hidde L. Ploegh (1,3 *) Stephanie K. Dougan (1,4), Robert A. Weinberg (1,3,5).
1. Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
2. Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA.
3. Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
4. Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
5. Ludwig Center for Molecular Oncology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
*Present address: Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA.
Tracking tumorigenesis

Elizabeth Li ’18 has worked in three cancer-related labs over the past six years, and one day intends to start her own.

Raleigh McElvery
March 6, 2018

Senior Elizabeth Li recreates miniature organs — lungs and intestines — outside the body. She does so in order to study cancer progression in both environments, and over the past six years has worked in three separate cancer-focused labs: two at MIT and another beginning her junior year of high school. One day, she aims to run her own.

“I’ve been into math and science ever since I was little,” she explains, “but in third grade I met a friend who was pretty important to me. She was diagnosed with a very malignant form of brain cancer and ended up dying from it. From that point on — even though I was still very young — I knew I wanted to do cancer research.”

In 9th grade, Li began at the School for Science and Math at Vanderbilt, a joint program between the university and Metro Nashville Public Schools. “I got to skip school once a week to learn research techniques, and had the opportunity to join Andries Zijlstra’s lab my junior year,” she recalls. “I’m actually still part of that group, and I’ve been working on the same project related to cancer metastasis for six years now.”

When it came time to select a university, Li was torn between Vanderbilt — where she was already performing research — and MIT, which she describes as “the place of opportunity.” She was ultimately swayed by MIT’s vast array of research areas, and fully sold after an overnight to preview the undergraduate culture.

Li opted for Course 7 in order to continue doing cancer research, and joined Omer Yilmaz’s lab in 2015 to investigate intestinal tumorigenesis. Here, she spent most of her time doing organoid work, studying the progression of colorectal cancer in miniaturized and simplified versions of the intestine. Li removed individual intestinal stem cells — or sometimes the entire “crypts” in which they reside — and grew them inside a 3D gel. This environment allowed the cells to differentiate and interact as they would in the colon, rather than growing on a flat, plastic dish.

Li and other members of the Yilmaz lab watched these cells multiply, observing their shape and the regeneration process. Li’s method of assessment varied depending on the research question: on some days, she stained the cells for proliferation markers, and on others she exposed them to different metabolites or drugs to see how the cells responded.

“On a typical day, I would come in during the morning between classes, and again in the afternoons and evenings,” she says. “The experiments differed, but we tended to do a lot of genetic manipulation. We’d make plasmids, CRISPR-Cas9 knockouts, or test for gene and protein expression using qPCR and Western blots.”

After two years, Li’s primary mentor finished her postdoctoral training, and Li transitioned to Jackie Lees’ lab at the beginning of her senior year. Li is now working with a fellow undergraduate on an independent project, centered on the enzyme protein arginine methyltransferase 5 (PRMT5).

PRMT5 catalyzes the transfer of methyl groups to the amino acid arginine in certain proteins, modifying their function. The enzyme also plays a key role in regulating gene splicing, the process by which segments of pre-mRNA are removed — changing the genetic code so that multiple genes can be encoded by the same initial transcript.

The Lees lab is interested in PRMT5 because it affects glioblastoma formation, the most common form of adult brain malignancy. As Li explains, when PRMT5 expression increases, so does tumor formation. Since there are still relatively few therapeutic options to treat glioblastomas, she’s hoping to use small molecules to inhibit PRMT5 expression and thus impede tumor initiation and progression.

“We’re considering using nanoparticles to deliver them,” she says, “and in doing so, hoping to gain a better understanding of how PRMT5 inhibition might impact cancer progression and tumorigenesis.”

Li is testing one small molecule PRMT5 inhibitor in lung organoids and several 2D cell lines — determining how sensitive the cells are and if the organoids will form, to gauge whether a tumor would still develop in the presence of the drug. “Depending on when you add the drug, you can test different aspects of tumorigenesis,” she explains.

She’s also split the past four years between the Biology Undergraduate Student Association (of which she was faculty liaison, outreach chair, and then co-president), the MIT Pre-Medical Society, MIT Lion DanceAsian Dance TeamWind Ensemble, and Improv-a-Do! She’s also heavily involved in DynaMIT, which organizes an annual, week-long science program for economically disadvantaged middle school students.

“There are a lot of extracurriculars to do,” Li admits. “But it’s pretty easy to get involved in the MIT community and still stay on top of your coursework, if you keep it to four or five classes per semester. It’s worked out for me so far — I’m still alive and happy and have time for eating, sleeping, and friends.”

As Li applies to MD-PhD programs, she hopes one day to practice medicine (perhaps pediatric oncology) while running her own lab.

“My advice for incoming MIT undergrads would be to remember to have fun,” she says. “You only have four years, so take advantage of your time here: hang out with your friends, take the classes you want to take, and do things that you enjoy. Hopefully most of those activities will be one and the same.”

Photo credit: Raleigh McElvery
The Man Who Uncovered a New (Old) Way to Fight Cancer

Matthew Vander Heiden helped revive the forgotten— but critical—study of cancer metabolism.

Sam Apple | MIT Technology Review
February 21, 2018

Person in plaid shirt by windowOne day last October, MIT biology professor Matthew Vander Heiden showed up in one of his trademark plaid shirts to teach his undergraduate course on cancer biology. As usual, he peppered his lecture with questions, filling six sliding chalkboards with arrows mapping cellular pathways; he had to erase the boards halfway through class to make room for more notations. But what might have seemed like an ordinary lecture was far from ordinary in one respect: although Vander Heiden was explaining some of the most fundamental aspects of how tumors grow, most of what he was teaching his students would have been absent from nearly every introductory course on cancer biology a decade ago. The science Vander Heiden discussed that afternoon amounted to a once-lost but recently rediscovered chapter in the history of cancer research.

What he didn’t mention in class is that he’d played as large a role as anyone in bringing it back.

That lost chapter focuses on metabolism, and how cancers use nutrients for energy and as building blocks for new cancer cells. It began with a discovery in the early 1920s that most cancers stuff themselves with glucose and then use it in an unusual way. Whereas normal cells typically break down glucose by burning it with oxygen, cancer cells extract much of its energy through fermentation—essentially the same process microorganisms use to make yogurt, beer, and other foods. Indeed, early-20th-century researchers noticed that cancer cells seemed to behave more like yeast than the cells of an animal. But though it would briefly become a major school of cancer research, metabolism fell by the wayside in the 1960s as researchers turned their attention to how cancer-causing genes signal cells to divide.

Cancer metabolism research appeared to be dead, until Vander Heiden helped launch its revival around two decades ago. Today it’s among the hottest areas of the field, spawning conferences, journals, and promising new therapies. And it has fundamentally changed the way many researchers understand cancer and its origins.

Modest revolutionary

Metabolism’s downfall as a research area in the late 20th century was largely a reflection of the faddish nature of science. It didn’t help that Otto Warburg, the German scientist who discovered the unusual metabolism of cancer cells, was so arrogant that much of the scientific community disliked him. So it’s probably a good thing that Vander Heiden, a down-to-earth type who’s been known to downplay his own role on research papers to give his students and postdocs first-author billing, has been so central to the metabolism revival.

Vander Heiden, 45, grew up in Port Washington, Wisconsin, a small town on Lake Michigan once known for its lawnmower factories, and he lives up to every stereotype of his background. “He carries his Midwestern sensibilities with him everywhere he goes,” says his wife, Brooke Bevis, a biologist and the operations manager for Vander Heiden’s lab at MIT’s Koch Institute for Integrative Cancer Research. “I finally made him give up my old 1995 Honda Civic just a few years ago.”

When Vander Heiden enrolled at the University of Chicago in 1990, medicine was already on his mind. His younger brother had suffered from a rare blood disorder as a child, and Vander Heiden spent much of his own childhood hanging around children’s hospitals. But he had little thought of becoming an academic scientist until he began a work-study job washing out equipment in a University of Chicago biology lab. The work was not glamorous but came with a perk: the graduate students in the lab would let Vander Heiden make solutions for them and show him how they did their experiments.

After graduating, he enrolled in Chicago’s MD-PhD program and landed in the lab of Craig B. Thompson. Today Thompson is the president and CEO of the Memorial Sloan Kettering Cancer Center, but at the time he was studying immunology, looking at how the body eliminated huge numbers of immune cells once they were no longer needed.

When Vander Heiden arrived in Thompson’s lab in 1996, part of the explanation was already understood. Those cells would simply commit suicide, a process known as apoptosis. It was also known that a family of proteins named Bcl-2 could stop a cell from committing suicide—and that they appeared to do so through their impact on mitochondria, tiny organelles known as the powerhouses of the cell for their role in energy production.

Vander Heiden had just joined a cutting–edge immunology lab interested in protein signaling. Yet he had been asked to investigate how Bcl-2 proteins affect mitochondria, a relic of the old, outdated metabolism research. When it became clear that no one in the lab knew much about metabolism, Vander Heiden reread the relevant sections of his undergraduate biochemistry textbook. He also teamed up with Navdeep Chandel, a metabolism researcher at Northwestern University who was then a graduate student in a University of Chicago cellular physiology lab.

When another lab showed that proteins released from the mitochondria could trigger apoptosis, Vander Heiden and -Chandel got an important clue: the decision to commit suicide could now be traced directly to the mitochondria. And yet the deeper question of what was happening inside them remained mysterious until the two researchers arrived at an answer, thanks to a series of elegant experiments designed by Vander Heiden (whom Chandel calls “a world-class experimentalist”) to study how molecules moved through the mitochondrial membrane. They discovered that the release of the mitochondrial proteins was a sign of a failing powerhouse, a notice to the cell that a brownout was under way so it was time to abort. But brownouts weren’t inevitable; the Bcl-2 proteins, like emergency workers called to the scene of an imminent disaster, could resuscitate the metabolic function of the mitochondria and keep things from getting to that point. The suicide signal, in turn, would never be released.

Person pipets in lab hood
Daniel Schmidt, a postdoc in Vander Heiden’s lab, prepares cells to study how metabolism affects cancer cell proliferation. Credit: BUCK SQUIBB
Daniel Schmidt, a postdoc in Vander Heiden’s lab, prepares cells to study how metabolism affects cancer cell proliferation.

BUCK SQUIBB

For Vander Heiden, this was a “watershed moment.” Among other things, it meant that metabolic enzymes weren’t merely supplying energy from food. Metabolism was governing the most fundamental decision a cell has to make—whether to live or die. That meant it had to be interwoven into the signaling cascades that molecular biologists studied. His feeling at the time, he recalls, was “Oh my goodness. We don’t really understand metabolism.”

Vander Heiden might not have envisioned himself delving into research areas that had been discarded decades earlier, but what was more surprising was how little research was then being done in an area that was “as fundamental as you get in terms of how biology works,” he says. “I looked around and no one was studying it.”

Thompson, recognizing the opportunity, shifted the focus of his lab to metabolism. Vander Heiden, meanwhile, continued to pursue Thompson’s broader question of how the body eliminates unwanted immune cells. He already knew that growth factors, messages sent from one cell to the next, kept cells from committing suicide, but how the signals delivered their survival message remained unclear. What he discovered in a series of studies carried out in the late ’90s followed perfectly from his previous research. Growth factors kept cells alive by giving them permission to eat. Without that permission, a cell soon faced an energy crisis, and the mitochondria released their death signals.

The takeaway was clear: our bodies eliminate unwanted cells by starving them to death.

Solving the metabolism mystery

As Vander Heiden’s MD-PhD program was coming to an end, he hadn’t yet begun to focus on cancer, but its possible links with his research on cell suicide were intriguing. Cancer cells were the other side of the coin—cells that were resistant to suicide, that no longer cared about instructions from other cells. So in 2004, after completing a residency in oncology at Brigham and Women’s Hospital in Boston, he was anxious to investigate cancer metabolism for his postdoc research.

Finding the right lab wasn’t easy. At the time, telling leading researchers he wanted to study how cancer cells consumed glucose was like approaching a high-tech manufacturer and announcing you wanted to study the trucks that brought fuel to the factory. It sounded, Vander Heiden says, “like a really ridiculous thing.”

Vander Heiden eventually found a home in the Harvard lab of Lewis Cantley, who now directs the Meyer Cancer Center at Weil Cornell. His research in Cantley’s lab would help solve one of the central riddles of cancer metabolism: why cancer cells are so ravenous for glucose. Researchers had once assumed that cancer cells were turning to fermentation because they’d lost the ability to use oxygen properly and needed some other way to produce energy. But Vander Heiden’s research on a mutated form of the enzyme pyruvate kinase showed something else. Rather than being used for energy, much of the glucose was being shunted into pathways used to build new molecules. What a growing cancer needs most of all from its food, the research suggested, is more spare parts—raw materials for making new DNA, membranes, and proteins.

Rethinking chemotherapy

Vander Heiden’s research with Cantley would also lead to his involvement with Agios Pharmaceuticals, the company behind one of the most promising new drugs to emerge from the metabolism revival. (Cantley says he played a major role in building the company’s science in its early days.) The drug, AG-221, treats acute myelogenous leukemia, a cancer of the blood and bone marrow. It works by blocking the product of a mutated form of the mitochondrial enzyme IDH-2. Approved by the US Food and Drug Administration in August, it has been hailed as the first real advance for the disease in 30 years.

The approval of AG-221 isn’t the only thing generating excitement in the cancer world. Unlike almost all other cancer drugs, AG-221 doesn’t kill the cancer cells but, rather, allows them to develop out of their deranged state into noncancerous, mature, functioning cells. That a single metabolic enzyme could have such profound effects on which genes are expressed in a cell is now one of the many signs that changes in metabolism are not just a response to the needs of a growing cancer. Often, they may actually be causing the cancer itself. It represents a major shift in thought: many cancer-causing genes long known for their ability to signal cells to keep dividing have now been shown to have additional roles in signaling cells to keep eating. Some researchers now believe the overeating typically comes first, driving the transformations that follow.

Since his arrival at MIT and the opening of his lab at the Koch Institute in 2009, Vander Heiden has treated cancer patients and continued to search for better therapies. In recent years he has focused on improving understanding of chemotherapy. Though typically thought of as general poisons, most chemotherapy drugs work because they disrupt metabolic functions. That much has long been known, but less clear is why a particular drug works for some cancers and not for others, even when two cancers carry the same mutations.

It was while explaining to his undergrad cancer biology students how targeted drugs work that Vander Heiden first thought of an answer. As a cancer doctor, he knew that chemotherapies are often chosen on the basis of where in the body a tumor first arose, but what was it about this location that made the difference?

Vander Heiden’s research in mice now suggests that the answer may lie in which foods are available to the cancer as it forms. Melanoma and colon cancer, for example, often have the same mutations, and yet, as he explains, because the two cancers “grow in very different places in the body,” they likely “have access to different nutrients.” He adds, “It has nothing to do with the genetics.” If he turns out to be right, it could lead to a fundamental change in how oncologists think about which drugs to give their patients.

As Vander Heiden turns his attention to old chemotherapy drugs, rethinking why and how they work, he is once again looking to the past for new insights on cancer. It might be more than a coincidence. As Bevis, his wife, says, the outdated Honda Civic isn’t the only item he has struggled to let go of. “The list goes on and on,” she says. “He hates waste and will use items long after someone else would have replaced them with a newer, shinier model.”

Probing a critical player in cancer growth

Alissandra Hillis ’18 has spent all four years at MIT in the same cancer metabolism lab, deciphering the basic science behind pancreatic cancer.

Raleigh McElvery
February 19, 2018

Senior Alissandra Hillis attributes her appetite for the basic sciences to her craving for fundamental knowledge. She’s spent her four years at MIT in the same lab, committed to unraveling the molecular mechanics of pancreatic cancer — the fourth leading cause of cancer death for both men and women, given that symptoms do not often appear until the disease is quite advanced.

“I was always very curious growing up,” she says. “I taught myself how to read at a very young age, just because I wanted to know about things and how they worked. But I didn’t become interested in biology and chemistry specifically until I came to MIT and started taking my General Institute Requirements.”

In doing so, Hillis became enthralled by the prospect of breaking down life into its most fundamental, biological units to decipher cellular function and disease. Originally a Course 7 major with a chemistry minor, she declared Course 5-7 (Chemistry and Biology) as soon as it became available in the fall of 2017 — applying her study of biochemistry and cell metabolism to cancer research.

“When I was quite young, my grandfather was diagnosed with stomach cancer, and ended up having almost three quarters of his stomach removed,” she says. “I was too little to really understand the severity of the situation, but as soon as I came to MIT I started to wonder what was going on at a cellular level. Most people today know someone who is fighting cancer, and yet we’re still lacking effective treatments for its most severe forms.”

Hillis joined Matthew Vander Heiden’s cancer metabolism lab the first semester of her freshman year, and has been there ever since.

Professor Vander Heiden does an excellent job of tailoring the research project to the individual, and there is no hierarchy among lab members,” she says. “I really liked it from the onset, so I stayed.”

For nearly two years, Hillis has been investigating the role of one enzyme, pyruvate kinase muscle isozyme M2 (PKM2), in pancreatic cancer. PKM2 is responsible for catalyzing the final step in glycolysis, which is required to create the energy that fuels cells. Glycolysis is also important in tumor metastasis and growth, since cancer cells demand energy in order to proliferate.

Cancer cells often preferentially express PKM2 over other types of pyruvate kinases such as PKM1. This spurred William Israelsen PhD ’14, a former graduate student in the Vander Heiden lab working in breast cancer models, to delete the PKM2 gene and see what happened. Since PKM2 is critical for glycolysis, and cancer cells require energy to proliferate, he anticipated that removing PKM2 would hinder energy production and thus disrupt tumor development. To his surprise, he found the opposite: deleting PKM2 actually accelerated tumor formation and promoted liver metastasis in mice.

In his 2014 paper, Israelsen concluded that PKM2 might permit cancer cells to maintain their “plasticity,” shifting from one specialized role to another even after they’ve fully matured. In the absence of PKM2, he proposed PKM1 might take over PKM2’s influential role.

Hillis wondered if she could replicate Israelsen’s breast cancer results in a model for pancreatic cancer, especially given the conflicting findings in human data regarding PKM2 expression in the latter. Some studies suggest that high PKM2 expression correlates with accelerated disease, whiles others indicate just the opposite: that high PKM2 expression is associated with better survival rates.

“Going into the project, we were expecting similar effects in both pancreatic and breast cancer models because both cancers preferentially express PKM2, and we were using the same method of PKM2 deletion, just bred into a different cancer model,” Hillis explains. “We anticipated that PKM2 deletion would accelerate pancreatic tumor size and tumor genesis, and decrease the mouse’s lifespan. But we’ve noticed that these effects — if they exist — are very much attenuated in the pancreatic cancer model; there is only a slight decrease in lifespan and increase in tumor size without PKM2.”

Right now, her working hypothesis holds that PKM2’s influence varies depending on the tissue in question. This might explain why her own results don’t exactly parallel what Israelsen found in his breast cancer model. For instance, the method they were using to delete PKM2 is quite effective in the breast and pancreatic cells themselves, but less so in the dense scar-like tissue characteristic of pancreatic tumors in particular. It’s possible, she thinks, that this fibrous tissue may still express some PKM2 even post-knockout, perhaps hindering both a drastic decrease in lifespan and increase in tumor size.

Hillis hopes piecing together PKM2’s mechanism of action will help us better diagnose — and eventually treat — certain cancers. Her most recent results were published in the November 2017 issue Cancer & Metabolism.

Although Hillis enjoys tackling the more fundamental questions concerning cancer, she’s also interested in translating this work from bench to bedside. That’s why she decided to intern with David Ting at the Massachusetts General Hospital Cancer Center this past summer.

“I wanted to try a different type of research before applying to graduate school,” she says. “The Department of Biology frequently sends out emails about job opportunities, and there was one advertising that the Ting lab was looking for a research technician.”

Although she was still a junior at the time, she contacted Ting — an MIT alumnus with a dual degree in 7A and 10 — and together they fashioned a summer position just for her, studying the role of miniscule, fluid-filled transportation structures called exosomes in cancer development and diagnosis.

“That was the first time I’d worked with samples from actual patients,” she says. “Many of the assays were the same, but I felt closer to a clinical application than I ever had before. I really enjoy doing the foundational work to identify the basic problem, but there’s definitely something to be said for experiencing research targeted at creating a diagnostic tool. I can see the pros and cons of both approaches.”

As Hillis begins her final semester at MIT, she’s continuing her work in the Vander Heiden lab, while also finishing up the requirements for her HASS concentration in legal studies. She’s still set on pursuing a PhD in cancer biology, but the propensity to ask tough questions that drew her to science in the first place has led her to realize that the questions she raises in her own research have ramifications far beyond her lab bench. Taking policy-oriented classes in addition to her science-related ones has inspired her to pursue a law degree in conjunction with her PhD — weaving together her love for science with a newfound interest in the rules and regulations that govern how science is funded, performed, shared, applied, and monetized.

“I really enjoy doing research, and that’s something I probably will continue to do,” she says, “but I also want to influence science-related regulations, which is something I couldn’t possibly do without a law degree. I would still be heavily immersed in science, while applying the subjects I love in new and exciting ways.”

Photo credit: Raleigh McElvery
Fine-tuning cancer medicine

New cancer research initiative eyes individualized treatment for patients.

Koch Institute
February 1, 2018

Details matter — perhaps most noticeably in the fight against cancer. Some patients respond to a given anticancer therapy, and some do not. A new initiative at MIT takes aim at those details, and the name of the game is precision.

The recently launched MIT Center for Precision Cancer Medicine (CPCM) is housed within MIT’s Koch Institute for Integrative Cancer Research and headed by physician-scientist Michael B. Yaffe, the David H. Koch Professor of Science and professor of biology and biological engineering. The center brings together leading Institute faculty members to focus on key research themes to accelerate the clinical translation of novel cancer discoveries, treatments, and technologies.

Engineering approaches to the clinic

While other institutions have begun efforts in precision medicine as well, the MIT Center for Precision Cancer Medicine stands out for using engineering approaches to solve complex clinical challenges in cancer treatment that are rooted in biology. In particular, the CPCM combines understandings of biological circuitry — along with engineering, computational, and mathematical techniques (as well as genomic ones) — to focus on signaling networks and pathways that are aberrantly regulated in cancer cells. This strategy is supported by the fact that most state-of-the-art molecularly targeted cancer therapies are focused on these key pathways.

At its core, the CPCM is driven by both internal and external collaboration, and is devoted to translational research to help the substantial number of patients who do not respond well to traditional cancer therapies — for example, those with triple-negative breast cancer, ovarian cancer, non-small cell lung cancer, or advanced prostate cancer.

To improve outcomes for these patients, CPCM investigators are focused on four key areas of research. First among these is identifying and targeting the processes, signals, and mechanisms that determine an individual patient’s response to chemotherapy. Recent discoveries by CPCM researchers include mechanisms that cancer cells use to repair chemotherapy damage that should have killed them, to hide from drugs in protected “niches” in the body, or to grow when and where they should not.

CPCM members are also working on a second research pillar, which involves finding ways to use existing FDA-approved cancer drugs more effectively, particularly in carefully designed combinations. Combination therapies are currently used in the clinic to treat some cancers, yet the discovery process for these has been largely empirical. By contrast, CPCM investigators are integrating their knowledge of cancer biology, understandings of drugs’ mechanisms of action, and sophisticated analytical techniques, to identify or design specific combinations that work synergistically to disarm and then destroy cancer cells.

“We believe we can significantly alter cancer patients’ outcomes by determining the right combination of therapies and the right sequence of drugs for the right patients,” says Yaffe. “We’re also concentrating on innovative ways to give these drugs, like time-staggered dosages and nanoparticle delivery.” He notes that, as part of their analyses of drugs and combination regimens currently administered in the clinic, CPCM members expect to identify combinations of drugs that are not as efficacious when given simultaneously as when given sequentially, at specific intervals. Yaffe stresses that these will be important findings that could help reduce the toxicity of treatment by not exposing people to multiple drug toxicities at the same time.

In parallel with their efforts to use existing drugs more effectively, CPCM investigators are also working to identify compounds, materials, and approaches that can engage key “undruggable” genetic and molecular targets and disrupt processes driving drug resistance. The “undruggable” label often refers to the fact that a target protein or molecule lacks a site to which drugs can bind, and thus is not considered a good drug target by the pharmaceutical industry. However, using novel chemistry approaches, CPCM researchers have made early inroads against several such high-value cancer targets, including specific transcription factors and RNA-binding proteins. The center will continue and expand these efforts as the third part of its research platform, including collaborations with industry.

Finally, the fourth component of the CPCM’s efforts will be harnessing MIT’s particular expertise in big data analysis and tools to begin new and expedite existing cancer research efforts. For example, the researchers plan to use data analytics to identify selective panels of biomarkers that can be used to prioritize which of their drug combinations, treatment protocols, and formulations are best suited to a particular patient’s tumor.

Getting discoveries out the door

“Patients will be the ultimate beneficiaries of the work of the new MIT Center for Precision Cancer Medicine,” says Tyler Jacks, director of the Koch Institute and the David H. Koch Professor of Biology. “This research is, by its nature, imminently and rapidly translatable. By concentrating efforts on which patients will benefit from particular existing drugs or combinations of drugs, there is a relatively small step from laboratory to a treatment that is benefitting a cancer patient.”

While work on combinations of approved therapies, like that at the CPCM, may be more rapidly translatable than other cancer research, it can be challenging for industry to pursue, particularly when those drugs hail from multiple companies. Overcoming this disjuncture is one of the goals behind the establishment of the MIT Center for Precision Cancer Medicine, which was made possible by a generous gift from an anonymous donor.

Yaffe and his CPCM colleagues are committed to finding viable routes to move their cancer research into the clinic, particularly through collaborations between CPCM members, hospitals, and industry. Logistically, this means more work for the center’s research groups, including advanced laboratory and preclinical studies, safety and scale-up studies, and clinical-grade manufacturing, as well as staff to carry it out. Woven into these efforts, CPCM investigators will tap into MIT’s celebrated tradition of entrepreneurship and, even more so, the Institute’s expanding network of clinical collaborators. The philanthropic investment behind the center will provide stable financial support for the researchers’ endeavors.

The new hub in town

In addition to supporting the research of member investigators, the CPCM offers a robust training ground for young engineers and scientists interested in precision medicine. Moreover, it will serve as the hub of precision cancer medicine research at MIT and beyond, connecting with researchers across the MIT campus and partnering with clinical investigators in Greater Boston’s noted health care centers and around the country.

Five outstanding cancer researchers make up the center’s founding faculty:

  • Michael B. Yaffe, MD, PhD, director, MIT Center for Precision Cancer Medicine; David H. Koch Professor of Science, professor of biology and biological engineering
  • Michael Hemann, PhD, associate professor of biology
  • Angela Koehler, PhD, Karl Van Tassel (1925) Career Development Associate Professor, assistant professor of biological engineering
  • Matthew Vander Heiden, MD, PhD, associate professor of biology, associate director, Koch Institute for Integrative Cancer Research
  • Forest M. White, PhD, professor of biological engineering

Efforts are currently underway to recruit an assistant director and a scientific advisory board.

As part of its charge, and key to spurring the new collaborations in precision cancer medicine that are its focus, the MIT Center for Precision Cancer Medicine will also convene lectures, events, and scientific exchanges and symposia, the first of which is slated for the fall.

December 14, 2017

Lab coat meets legislation

Person with red hair in bun stands outside with umbrella.

Undergraduate Courtney Diamond combines biology and policy to tackle real-world challenges

Raleigh McElvery

 

Undergraduate Courtney Diamond arrived at MIT determined to be an oncologist. Five years later, she’s leaving with a broader focus on human health, grappling with real-world, biomedical problems by way of public policy rather than medicine or research.

Although Diamond had completed her requirements for a degree in biology at the beginning of her senior year, she decided then to add a second major: Course 17 (Political Science), and with it a fifth year of study at MIT.

“I came into MIT wanting to be a doctor, but the more I thought about it the less it felt like medical school would be a good fit,” she says. “I spent a long time narrowing my interests within the realm of human health, and recently realized there was another dimension to that interest related to public policy, which was also this common thread among my extracurriculars.”

Diamond grew up in a small town in Massachusetts called Millbury, not too far from MIT, which she describes as special to her but “rather unremarkable” in most other ways — with the exception of one particularly zealous and articulate high school biology teacher. His infectious enthusiasm sparked Diamond’s passion for the life sciences, but over the course of her senior year this interest became far more personal. It was around that time that her mother developed breast cancer, and Diamond resolved to be an oncologist.

“My mom had been diagnosed once before with a different kind of cancer, cervical cancer,” she says. “But I was in sixth grade back then, and assumed she was just at home resting. By the time the breast cancer rolled around, I was old enough to understand that most people are lucky to survive cancer once. But twice?”

Her mother has since entered remission, and the year Diamond began at MIT her interests matured away from a career in medicine and towards biomedical research. In April 2014, she applied to the MIT Undergraduate Research Opportunities Program (UROP). “I wanted to figure out which part of biology excited me — which area I really wanted to drill down on,” she recalls.

She began working with a postdoctoral fellow in Professor Darrell Irvine’s lab at the Koch Institute for Integrative Cancer Research, tackling research questions related to cancer immunology. Diamond’s job was to analyze murine tumors as they developed over time, in order to understand how they were affected by changes to their cellular environments.

After a year, Diamond took a break from research in order to focus on her classes. But she didn’t stay away for long.

“I’ve had a life-long obsession with Australia,” she says, “and in the fall of my sophomore year, I told my advisor, Professor Bob Horvitz, that my dream was to study biochemistry in Melbourne.” One email and two hours later, she received an offer from the Walter and Eliza Hall Institute for Medical Research to spend a summer abroad in Jeff Babon’s lab. “It turns out the director of the Institute did his postdoc at MIT, and liked the UROP system so much he decided to bring it back to Australia,” she explains.

There, Diamond helped to unravel the structure of a protein complex known as JAK-STAT. This complex is involved in many diverse processes — from cell proliferation and programmed cell death to immunity — making it critical to understand how the different molecular components of the complex fit together to influence function.

When she returned to MIT, Diamond decided to maintain her focus on structural biology. She completed her thesis in Professor Thomas Schwartz’s lab, studying the Y complex, a component of the nuclear pore — a channel that allows mRNA and other molecules to pass into the cell’s nucleus. Diamond helped creat a library of fluorescing antibodies that could adhere to the Y complex, allowing her to visualize its position within the nuclear pore. After a year, she opted to broaden her interests by taking classes outside her major.

One of those classes, recommended by a friend, was in political science: 17.309 (Science, Technology, and Public Policy), taught by Professor Kenneth Oye. During one of his lectures, Oye made a quip about a small Massachusetts town called Millbury.

“I came up to him after class to ask him, ‘Did you know I’m actually from there?’ and he thought it was the funniest thing,” she says. “That initial, informal interaction led to more meaningful conversations, and I ended up working with him on a few projects.”

Today, she is pursuing a final UROP with Oye, looking at technologies and policies related to synthetic biology. At Oye’s weekly working group of graduate students and postdocs, she debates the possible repercussions of using gene editing techniques like CRISPR-Cas9 to control the transmission of certain traits throughout a given population. For example, what would happen if mosquitos in the regions where malaria is most prevalent carried a gene encoding malaria resistance — would that eradicate the illness? But might there be unintended, negative consequences?

As part of a separate project, Diamond is researching U.S. consent and privacy policies in the realm of health information technology. She’s also hard at work on her political science thesis, focusing on ways to incentivize companies and researchers to develop new and more effective antibiotics to combat antimicrobial resistance.

Diamond is now applying for public health consulting jobs, and she plans to pursue graduate training in epidemiology, followed by a master’s in public health. Long-term, she sees herself at the Centers for Disease Control and Prevention or the World Health Organization.

“I mean, that’s the current plan,” she says. “Check back in with me in two years.”

Photo credit: Raleigh McElvery
From DNA forensics to cancer metabolism

Carolyn Lanzkron discovered bench science while attending community college with her son, and followed her newfound passion to MIT

Raleigh McElvery
December 3, 2017

From DNA forensics to cancer metabolism

Person in black hat and purple shirt sitting in front of lab building.

Carolyn Lanzkron discovered bench science while attending community college with her son, and followed her newfound passion to MIT

Raleigh McElvery

 

Carolyn Lanzkron spent 20 years as a stay-at-home mother raising five children before starting at MIT. Life has taught her patience, which she, in turn, has tried to pass on to her kids: “A successful person falls down many times and gets up — just pick a direction and move forward.”

Those were the same words she told her teenage son back in 2011 when she encouraged him to attend community college.

“I figured I would just take a few courses with him,” she says. “He enjoyed his chemistry classes, so I was looking at the chemistry offerings, and on the wall there was a poster for Dr. Bruce Jackson’s unique Forensic DNA Science program.” Lanzkron was intrigued, and decided to enroll.

The students aided Jackson with real cases, and were given dedicated lab space and materials to follow their curiosities, as well as design their own inquiries. The program was based on a peer-mentoring model, and Lanzkron was appointed chief of peer mentors and forensic case manager. Under Jackson’s tutelage, she worked on lineage cases tracing ancestry and criminal cases for defense and prosecution.

“I was hoping my son would join me in a chemistry class, but he wasn’t so interested in having his mom as a lab partner — go figure,” she says. “But we carpooled to school together for a year, and by that time I’d developed a love for bench science.”

After two years, Lanzkron had completed her degree, but it wasn’t enough. So she applied to several institutions within her carpool radius, including MIT. Like all transfers here, she began as a sophomore.

“I love bench science because I really appreciate the combination of being part of a team and solving a big, important question, but at the same time having tasks in my day that allow me to focus on small details — like keeping track of the labels on my tubes,” she says. “That balance works really well for me; it satisfies my need for a quest while still having control over a small environment.”

She’s turned her attention from DNA forensics to cancer metabolism, an interest which has become far more personal over the past year. Last spring, Lanzkron’s mother was diagnosed with lung cancer, and Lanzkron took a leave of absence to care for her.

“Right now, my mother is doing really well, and we are enjoying a window of stability,” Lanzkron says, “which has allowed me to come back to MIT and finish my degree.”

Although Lanzkron is not currently in a lab, lest that period of stability suddenly end, she’s worked in several over the course of her three years at MIT. She began in Jean Francois Hamel’s chemical engineering lab, adapting an adherent cell line to grow in a suspension-like culture in various bioreactors using microcarriers.

Later, Lanzkron joined David Sabatini’s lab in the Whitehead Institute for Biomedical Research, aiding two separate projects: one spearheaded by then-postdoc Yoav Shaul, and the other led by MD-PhD student Walter Chen.

Chen was hard at work developing a new method for profiling undamaged mitochondria, while Shaul had discovered a unique set of 44 metabolic genes that were upregulated in certain cancers that expressed mesenchymal markers (which he called the “Mesenchymal Metabolic Signature,” or “MMS”), indicating that those cells were acquiring cancerous characteristics. Lanzkron collaborated with Shaul as he worked to further characterize the metabolic requirements and behavior of the MMS. She also helped him refine his web-based gene analysis tool, Metabolic gEne RApid Visualizer (MERAV), which queries a database comprising ∼4,400 microarrays, representing human gene expression in normal tissues, cancer cell lines, and primary tumors.

The summer after Shaul completed his postdoctoral training, Lanzkron interned in his lab in at the Hebrew University of Jerusalem at Hadassah Ein Kerem through the MISTI/Israel program, to continue working with him on these projects.

“When I went to Israel, my husband stayed in Boston and took care of the kids,” she recalls. “Without family responsibilities, I could work in lab around the clock, and that was great. I was actually able to finish things up, prepare them for the next day, and cover for other people and really focus; I look forward to being able to do that again as the kids get older.”

Lanzkron admits these aren’t the only aspects of the MIT undergraduate experience she’s missed — not just because she lives off campus and can’t meet at odd hours of the night to collaborate on problem sets — but also because she’s a generation and a half older than her classmates.

But in some ways she considers this an advantage. For instance, she now has the tools to guide her own children through today’s college process.

“I no longer have this outdated view of what it’s like to apply to schools and navigate the SAT,” she says. “Granted, MIT is not your average school. It’s been quite the ride to be at the community college where I had to bring my own masking tape to complete the gel trays because we didn’t have any sealing rings — I didn’t even know there was such a thing as a seal back then. And to go from that to the MIT Department of Biology and the Whitehead Institute where the resources are phenomenal, it’s just mind blowing. I have learned so much from both situations — having to make do, and having an abundance of resources.”

While Lanzkron intends to graduate this spring, her future plans depend on her mother’s health.

“I picked my classes this semester so that I could take her to her cancer treatment,” Lanzkron says, “so, though I’m ultimately planning to go to graduate school, right now things are still in flux.”

While maintaining this school-family balance would be inconceivable for most, Lanzkron takes her personal and academic responsibilities in stride.

“Honestly I’m so happy here at MIT,” she says. “I tell my kids, ‘Don’t get too worked up about the college process. You’ll get where you need to go — the starting point almost doesn’t matter; what matters is what you do when you get there.’”

Photo credit: Raleigh McElvery