CHAPTER 8

The Runaway Cell

New Ways to Address the Killer That Is Cancer

You may have to fight a battle more than once to win it.

—Margaret Thatcher

Steve Rosenberg was still a young resident when he encountered the patient who would determine the course of his career—and, possibly, of cancer treatment in general. He was on a rotation at a VA hospital in Massachusetts in 1968 when a man in his sixties came in needing a relatively simple gallbladder operation. The man, whose name was James DeAngelo, already had a large scar across his abdomen, which he said was from a long-ago operation to remove a stomach tumor. He had also had metastatic tumors that had spread to his liver, he added, but the surgeons had not touched those.

Rosenberg was sure that his patient was confused. It would have been a miracle if he had survived even six months with metastatic stomach cancer. But according to DeAngelo’s hospital records, that was exactly what had happened. Twelve years earlier, he had walked into the same hospital complaining of malaise and low energy. At the time, his chart noted, he was drinking his way through three or four bottles of whiskey per week and smoking a pack or two of cigarettes every day. Surgeons had discovered a fist-sized tumor in his stomach and smaller metastatic tumors in his liver. They removed the stomach tumor, along with half of his stomach, but they left the liver tumors alone, deciding that it was too risky to try to remove them at the same time. And then they had sewn him up and sent him home to die, which he had obviously failed to do.

Rosenberg went ahead with the gallbladder operation, and while he was in there he decided to take a look around in DeAngelo’s abdomen. He felt behind the liver, gingerly working his way under its soft purple lobes, expecting to feel lumps of remnant tumors—an unmistakable feeling, hard and round, almost alien-like—but he found absolutely no trace of any growths. “This man had had a virulent and untreatable cancer that should have killed him quickly,” Rosenberg wrote in his 1992 book The Transformed Cell. “He had received no treatment whatsoever for his disease from us or from anyone else. And he had been cured.”

How could this be? In all the medical literature, Rosenberg could find only four instances of complete and spontaneous remission of metastatic stomach cancer. He was mystified. But he eventually came up with a hypothesis: he believed that DeAngelo’s own immune system had fought off the cancer and killed the remaining tumors in his liver, the way you or I might shake off a cold. His own body had cured his cancer. Somehow.

At the time, this notion was well out of the mainstream of cancer research. But Rosenberg suspected that he was onto something important. The Transformed Cell told the story of Rosenberg’s quest to harness the immune system to fight cancer. Despite small successes peppered here and there, however, whatever phenomenon had erased James DeAngelo’s tumors proved to be elusive; for the first ten years, not a single one of Rosenberg’s patients had survived. Not one. But still he kept at it.

He was doing better as a cancer surgeon than a cancer researcher: he operated on President Ronald Reagan in 1985, removing cancerous polyps from his colon, and that had gone fine. But Rosenberg’s goal was to eliminate the need for cancer surgeries, period. Finally, in the mid-1980s, he had a glimmer of success—just enough to keep him going.


As soon as I read The Transformed Cell, as a medical student, I knew that I wanted to be a surgical oncologist and that I had to work with Steve Rosenberg. Cancer had been on my mind since before I had even applied to medical school. During my postbac year, while taking med school prerequisite courses, I volunteered on the pediatric cancer ward of Kingston General Hospital in Ontario, spending time with kids who were undergoing cancer treatment. Thankfully, childhood leukemia is one area where Medicine 2.0 has made real progress. But not all the kids survived, and the bravery of these children, the pain that they and their parents endured, and the compassion of their medical teams moved me more deeply than any engineering or mathematical problem. It confirmed my decision to switch from engineering to medicine.

In my third year of medical school, I got the opportunity to spend four months in Rosenberg’s lab, at the epicenter of American cancer research. By the time I arrived, it had been nearly three decades since Richard Nixon had declared a national War on Cancer in 1971. Initially, the hope was that cancer would be “cured” within five years, in time for the Bicentennial. Yet it remained stubbornly undefeated in 1976, and still by the time I finished medical school in 2001. And today, for all intents and purposes.

Source: National Cancer Institute (2021).

Despite well over $100 billion spent on research via the National Cancer Institute, plus many billions more from private industry and public charities—despite all the pink ribbons and yellow bracelets, and literally millions of published papers on the PubMed database—cancer is the second leading cause of death in the United States, right behind heart disease. Together, these two conditions account for almost one in every two American deaths. The difference is that we understand the genesis and progression of heart disease fairly well, and we have some effective tools with which to prevent and treat it. As a result, mortality rates from cardiovascular disease and cerebrovascular disease have dropped by two-thirds since the middle of the twentieth century. But cancer still kills Americans at almost exactly the same rate as it did fifty years ago.

We have made some progress against a few specific cancers, notably leukemia (especially childhood leukemia, as I noted earlier). For adults with leukemia, ten-year survival rates nearly doubled between 1975 and 2000, leaping from 23 percent to 44 percent. Survival rates for Hodgkin’s and non-Hodgkin’s lymphomas have increased as well, especially the former. Yet these represent relatively small victories in a “war” that has not gone particularly well.

Like heart disease, cancer is a disease of aging. That is, it becomes exponentially more prevalent with each decade of life, as figure 6 shows. But it can be deadly at almost any age, especially middle age. The median age of a cancer diagnosis is sixty-six, but in 2017 there were more cancer deaths among people between forty-five and sixty-four than from heart disease, liver disease, and stroke combined. This year, if recent trends continue, that same age group will also account for nearly 40 percent of the estimated 1.7 million new cases of cancer that are likely to be diagnosed in the United States, according to the National Cancer Institute. By the time cancer is detected, however, it has probably already been progressing for years and possibly decades. As I write these words, I reflect sadly on three of my friends from high school who died of cancer in the past ten years, all younger than forty-five. I was able to say goodbye to only one of them before she died. Everyone reading this book probably has a few similar stories.

The problem we face is that once cancer is established, we lack highly effective treatments for it. Our toolbox is limited. Many (though not all) solid tumors can be removed surgically, a tactic that dates back to ancient Egypt. Combining surgery and radiation therapy is pretty effective against most local, solid-tumor cancers. But while we’ve gotten fairly good at this approach, we have essentially maxed out our ability to treat cancers this way. We are not getting any more juice from the squeeze. And surgery is of limited value when cancer has metastasized, or spread. Metastatic cancers can be slowed by chemotherapy, but they virtually always come back, often more resistant to treatment than ever. Our benchmark for success in a patient, or remission, is typically five-year survival, nothing more. We don’t dare utter the word cure.

The second problem is that our ability to detect cancer at an early stage remains very weak. Far too often, we discover tumors only when they cause other symptoms, by which point they are often too locally advanced to be removed—or worse, the cancer has already spread to other parts of the body. I saw this happen many times during my training: we would remove a patient’s tumor (or tumors), only to have them die a year later because that same cancer had taken hold elsewhere, like their liver or their lungs.

This experience informs our three-part strategy for dealing with cancer. Our first and most obvious wish is to avoid getting cancer at all, like the centenarians—in other words, prevention. But cancer prevention is tricky, because we do not yet fully understand what drives the initiation and progression of the disease with the same resolution that we have for atherosclerosis. Further, plain bad luck seems to play a major role in this largely stochastic process. But we do have some clues, which is what we’ll talk about in the next two sections.

Next is the use of newer and smarter treatments targeting cancer’s manifold weaknesses, including the insatiable metabolic hunger of fast-growing cancer cells and their vulnerability to new immune-based therapies, the outcome of decades of work by scientists like Steve Rosenberg. I feel that immunotherapy, in particular, has enormous promise.

Third, and perhaps most importantly, we need to try to detect cancer as early as possible so that our treatments can be deployed more effectively. I advocate early, aggressive, and broad screening for my patients—such as colonoscopy (or other colorectal cancer screening) at age forty, as opposed to the standard recommendation of forty-five or fifty—because the evidence is overwhelming that it’s much easier to deal with most cancers in their early stages. I am also cautiously optimistic about pairing these tried-and-true staples of cancer screening with emerging methods, such as “liquid biopsies,” which can detect trace amounts of cancer-cell DNA via a simple blood test.

Five decades into the war on cancer, it seems clear that no single “cure” is likely to be forthcoming. Rather, our best hope likely lies in figuring out better ways to attack cancer on all three of these fronts: prevention, more targeted and effective treatments, and comprehensive and accurate early detection.

What Is Cancer?

One major reason why cancer is so deadly—and so scary—is that we still know relatively little about how it begins and why it spreads.

Cancer cells are different from normal cells in two important ways. Contrary to popular belief, cancer cells don’t grow faster than their noncancerous counterparts; they just don’t stop growing when they are supposed to. For some reason, they stop listening to the body’s signals that tell them when to grow and when to stop growing. This process is thought to begin when normal cells acquire certain genetic mutations. For example, a gene called PTEN, which normally stops cells from growing or dividing (and eventually becoming tumors), is often mutated or “lost” in people with cancer, including about 31 percent of men with prostate cancer and 70 percent of men with advanced prostate cancer. Such “tumor suppressor” genes are critically important to our understanding of the disease.

The second property that defines cancer cells is their ability to travel from one part of the body to a distant site where they should not be. This is called metastasis, and it is what enables a cancerous cell in the breast to spread to the lung. This spreading is what turns a cancer from a local, manageable problem to a fatal, systemic disease.

Beyond these two common properties, however, the similarities among different cancers largely end. One of the biggest obstacles to a “cure” is the fact that cancer is not one single, simple, straightforward disease, but a condition with mind-boggling complexity.

About two decades ago, the National Cancer Institute launched a huge and ambitious study called The Cancer Genome Atlas, whose goal was to sequence cancer tumor cells in hopes of finding the precise genetic changes that cause various types of cancer, such as breast, kidney, and liver cancer. Armed with this knowledge, scientists would be able to develop therapies targeted at these exact mutations. As one of the scientists who proposed the project said, “These are the starting blocks that we need to develop a cure.”

But the early results of The Cancer Genome Atlas, published in a series of papers beginning in 2008, revealed more confusion than clarity. Rather than uncovering a definite pattern of genetic changes driving each type of cancer, the study found enormous complexity. Each tumor had more than one hundred different mutations, on average, and those mutations almost appeared to be random. A handful of genes emerged as drivers, including TP53 (also known as p53, found in half of all cancers), KRAS (common in pancreatic cancer), PIC3A (common in breast cancer), and BRAF (common in melanoma), but very few if any of these well-known mutations were shared across all tumors. In fact, there didn’t seem to be any individual genes that “caused” cancer at all; instead, it seemed to be random somatic mutations that combined to cause cancers. So not only is breast cancer genetically distinct from colon cancer (as the researchers expected), but no two breast cancer tumors are very much alike. If two women have breast cancer, at the same stage, their tumor genomes are likely to be very different from each other. Therefore, it would be difficult if not impossible to devise one treatment for both women based on the genetic profile of their tumors. Rather than revealing the shape of the forest, then, The Cancer Genome Atlas merely dragged us deeper into the maze of the trees.

Or so it seemed at the time. Ultimately, genome sequencing has proved to be a very powerful tool against cancer—just not in the way that was envisioned two decades ago.


Even when we treat a local cancer successfully, we can never be sure that it’s entirely gone. We have no way of knowing whether cancer cells may have already spread and are lurking in other organs, waiting to establish a foothold there. It is this metastatic cancer that is responsible for most cancer deaths. If we want to reduce cancer mortality by a significant amount, we must do a better job of preventing, detecting, and treating metastatic cancers.

With a few exceptions, such as glioblastoma or other aggressive brain tumors, as well as certain lung and liver cancers, solid organ tumors typically kill you only when they spread to other organs. Breast cancer kills only when it becomes metastatic. Prostate cancer kills only when it becomes metastatic. You could live without either of those organs. So when you hear the sad story of someone dying from breast or prostate cancer, or even pancreatic or colon cancer, they died because the cancer spread to other, more critical organs such as the brain, the lungs, the liver, and bones. When cancer reaches those places, survival rates drop precipitously.

But what causes cancer to spread? We don’t really know, and we are unlikely to find out anytime soon because only about 5 to 8 percent of US cancer research funding goes to the study of metastasis. Our ability to detect cancer metastasis is also very poor, although I do believe we are on the verge of some key breakthroughs in cancer screening, as we’ll discuss later. Most of our energy has been focused on treating metastatic cancer, which is an extremely difficult problem. Once cancer has spread, the entire game changes: we need to treat it systemically rather than locally.

Right now, this usually means chemotherapy. Contrary to popular belief, killing cancer cells is actually pretty easy. I’ve got a dozen potential chemotherapy agents in my garage and under my kitchen sink. Their labels identify them as glass cleaner or drain openers, but they would easily kill cancer cells too. The problem, of course, is that these poisons will also slaughter every normal cell in between, likely killing the patient in the process. The game is won by killing cancers while sparing the normal cells. Selective killing is the key.

Traditional chemotherapy occupies a fuzzy region between poison and medicine; the mustard gas used as a weapon during World War I was a direct precursor to some of the earliest chemotherapy agents, some of which are still in use. These drugs attack the replicative cycle of cells, and because cancer cells are rapidly dividing, the chemo agents harm them more severely than normal cells. But many important noncancerous cells are also dividing frequently, such as those in the lining of the mouth and gut, the hair follicles, and the nails, which is why typical chemotherapy agents cause side effects like hair loss and gastrointestinal misery. Meanwhile, as cancer researcher Robert Gatenby points out, those cancer cells that do manage to survive chemotherapy often end up acquiring mutations that make them stronger, like cockroaches that develop resistance to insecticides.

The side effects of chemo might seem at the outset to be a fair trade for a “chance for a few more useful years,” as the late author Christopher Hitchens noted in his cancer memoir Mortality. But as his treatment for metastatic esophageal cancer dragged on, he changed his mind. “I lay for days on end, trying in vain to postpone the moment when I would have to swallow. Every time I did swallow, a hellish tide of pain would flow up my throat, culminating in what felt like a mule kick in the small of my back….And then I had an unprompted rogue thought: If I had been told about all this in advance, would I have opted for the treatment?”

Hitchens was experiencing the primary flaw of modern chemotherapy: It is systemic, but still not specific enough to target only cancerous cells and not normal healthy cells. Hence the horrible side effects he suffered. Ultimately, successful treatments will need to be both systemic and specific to a particular cancer type. They will be able to exploit some weakness that is unique to cancer cells, while largely sparing normal cells (and, obviously, the patient). But what might those weaknesses be?

Just because cancer is powerful does not mean it is invincible. In 2011, two leading cancer researchers named Douglas Hanahan and Robert Weinberg identified two key hallmarks of cancer that may lead—and in fact have led—to new treatments, as well as potential methods of reducing cancer risk. The first such hallmark is the fact that many cancer cells have an altered metabolism, consuming huge amounts of glucose. Second, cancer cells seem to have an uncanny ability to evade the immune system, which normally hunts down damaged and dangerous cells—such as cancerous cells—and targets them for destruction. This second problem is the one that Steve Rosenberg and others have been trying to solve for decades.

Metabolism and immune surveillance excite me because they are both systemic, a necessary condition for any new treatment to combat metastatic cancers. They both exploit features of cancer that are potentially more specific to tumors than simply runaway cell replication. But neither the metabolic nor the immune-based approaches to cancer are exactly new: dogged researchers have been laying the groundwork for progress in both of these areas for decades.

Cancer Metabolism

As you might have gathered by now, we tend to think of cancer as primarily a genetic disease, driven by mutations of unknown cause. Clearly, cancer cells are genetically distinct from normal human cells. But for the last century or so, a handful of researchers have been investigating another unique property of cancer cells, and that is their metabolism.

In the 1920s, a German physiologist named Otto Warburg discovered that cancer cells had a strangely gluttonous appetite for glucose, devouring it at up to forty times the rate of healthy tissues. But these cancer cells weren’t “respiring” the way normal cells do, consuming oxygen and producing lots of ATP, the energy currency of the cell, via the mitochondria. Rather, they appeared to be using a different pathway that cells normally use to produce energy under anaerobic conditions, meaning without sufficient oxygen, such as when we are sprinting. The strange thing was that these cancer cells were resorting to this inefficient metabolic pathway despite having plenty of oxygen available to them.

This struck Warburg as a very strange choice. In normal aerobic respiration, a cell can turn one molecule of glucose into as many as thirty-six units of ATP. But under anaerobic conditions, that same amount of glucose yields only two net units of ATP. This phenomenon was dubbed the Warburg effect, and even today, one way to locate potential tumors is by injecting the patient with radioactively labeled glucose and then doing a PET scan to see where most of the glucose is migrating. Areas with abnormally high glucose concentrations indicate the possible presence of a tumor.

Warburg was awarded the Nobel Prize in Physiology or Medicine in 1931 for his discovery of a crucial enzyme in the electron transport chain (a key mechanism for producing energy in the cell). By the time he died in 1970, the weird quirk of cancer metabolism that he had discovered had been all but forgotten. The discovery of the structure of DNA by James Watson, Francis Crick, Maurice Wilkins, and Rosalind Franklin in 1953 had caused a seismic paradigm shift, not just in cancer research but in biology in general.

As Watson recounted in a 2009 New York Times op-ed: “In the late 1940s, when I was working toward my doctorate, the top dogs of biology were its biochemists who were trying to discover how the intermediary molecules of metabolism were made and broken down. After my colleagues and I discovered the double helix of DNA, biology’s top dogs then became its molecular biologists, whose primary role was finding out how the information encoded by DNA sequences was used to make the nucleic acid and protein components of cells.”

Nearly forty years into the War on Cancer, however, Watson himself had become convinced that genetics did not hold the key to successful cancer treatment after all. “We may have to turn our main research focus away from decoding the genetic instructions behind cancer and toward understanding the chemical reactions within cancer cells,” he wrote. It was time, he argued, to start looking at therapies that targeted cancer’s metabolism as well as its genetics.

A handful of scientists had been pursuing the metabolic aspects of cancer all along. Lew Cantley, now at Harvard’s Dana-Farber Cancer Center, has been investigating cancer metabolism since the 1980s, when the idea was unfashionable. One of the more puzzling questions he has tackled was why cancer cells needed to produce energy in this highly inefficient way. Because the inefficiency of the Warburg effect may be the point, as Cantley, Matthew Vander Heiden, and Craig Thompson argued in a 2009 paper. While it may not yield much in the way of energy, they found, the Warburg effect generates lots of by-products, such as lactate, a substance that is also produced during intense exercise. In fact, turning glucose into lactate creates so many extra molecules that the authors argued that the relatively small amount of energy it produces may actually be the “by-product.”

There’s a logic to this seeming madness: when a cell divides, it doesn’t simply split into two smaller cells. The process requires not only the division of the nucleus, and all that stuff we learned in high school biology, but the actual physical materials required to construct a whole new cell. Those don’t just appear out of nowhere. Normal aerobic cellular respiration produces only energy, in the form of ATP, plus water and carbon dioxide, which aren’t much use as building materials (also, we exhale the latter two). The Warburg effect, also known as anaerobic glycolysis, turns the same amount of glucose into a little bit of energy and a whole lot of chemical building blocks—which are then used to build new cells rapidly. Thus, the Warburg effect is how cancer cells fuel their own proliferation. But it also represents a potential vulnerability in cancer’s armor.[*1]

This remains a controversial view in mainstream cancer circles, but it has gotten harder and harder to ignore the link between cancer and metabolic dysfunction. In the 1990s and early 2000s, as rates of smoking and smoking-related cancers declined, a new threat emerged to take the place of tobacco smoke. Obesity and type 2 diabetes were snowballing into national and then global epidemics, and they seemed to be driving increased risk for many types of cancers, including esophageal, liver, and pancreatic cancer. The American Cancer Society reports that excess weight is a leading risk factor for both cancer cases and deaths, second only to smoking.

Globally, about 12 to 13 percent of all cancer cases are thought to be attributable to obesity. Obesity itself is strongly associated with thirteen different types of cancers, including pancreatic, esophageal, renal, ovarian, and breast cancers, as well as multiple myeloma (see figure 7). Type 2 diabetes also increases the risk of certain cancers, by as much as double in some cases (such as pancreatic and endometrial cancers). And extreme obesity (BMI ≥ 40) is associated with a 52 percent greater risk of death from all cancers in men, and 62 percent in women.

I suspect that the association between obesity, diabetes, and cancer is primarily driven by inflammation and growth factors such as insulin. Obesity, especially when accompanied by accumulation of visceral fat (and other fat outside of subcutaneous storage depots), helps promote inflammation, as dying fat cells secrete an array of inflammatory cytokines into the circulation (see figure 4 in chapter 6). This chronic inflammation helps create an environment that could induce cells to become cancerous. It also contributes to the development of insulin resistance, causing insulin levels to creep upwards—and, as we’ll see shortly, insulin itself is a bad actor in cancer metabolism.

This insight comes courtesy of further work by Lew Cantley. He and his colleagues discovered a family of enzymes called PI3-kinases, or PI3K, that play a major role in fueling the Warburg effect by speeding up glucose uptake into the cell. In effect, PI3K helps to open a gate in the cell wall, allowing glucose to flood in to fuel its growth. Cancer cells possess specific mutations that turn up PI3K activity while shutting down the tumor-suppressing protein PTEN, which we talked about earlier in this chapter. When PI3K is activated by insulin and IGF-1, the insulin-like growth factor, the cell is able to devour glucose at a great rate to fuel its growth. Thus, insulin acts as a kind of cancer enabler, accelerating its growth.

Source: NCI (2022a).

This in turn suggests that metabolic therapies, including dietary manipulations that lower insulin levels, could potentially help slow the growth of some cancers and reduce cancer risk. There is already some evidence that tinkering with metabolism can affect cancer rates. As we have seen, laboratory animals on calorically restricted (CR) diets tend to die from cancer at far lower rates than control animals on an ad libitum (all-they-can-eat) diet. Eating less appears to give them some degree of protection. The same may hold true in people: one study of caloric restriction in humans found that limiting caloric intake directly turns down the PI3K-associated pathway, albeit in muscle (which is not susceptible to cancer). This may be a function of lowered insulin rather than lower glucose levels.

While it’s tricky to impossible to avoid or prevent the genetic mutations that help give rise to cancer, it is relatively easy to address the metabolic factors that feed it. I’m not suggesting that it’s possible to “starve” cancer or that any particular diet will magically make cancer go away; cancer cells always seem to be able to obtain the energy supply they need. What I am saying is that we don’t want to be anywhere on that spectrum of insulin resistance to type 2 diabetes, where our cancer risk is clearly elevated. To me, this is the low-hanging fruit of cancer prevention, right up there with quitting smoking. Getting our metabolic health in order is essential to our anticancer strategy. In the next section, we’ll look at how metabolic interventions have also been used to potentiate other kinds of cancer therapy.

New Treatments

Lew Cantley’s discovery of the PI3K pathway led to the development of a whole class of drugs that target cancer metabolism. Three of these drugs, known as PI3K inhibitors, have been FDA approved for certain relapsed leukemias and lymphomas, and a fourth was approved in late 2019 for breast cancer. But they haven’t seemed to work as well as predicted, based on PI3K’s prominent role in the growth pathways of cancer cells. Also, they had the annoying side effect of raising blood glucose, which in turn provoked a jump in insulin levels and IGF-1 as the cell tried to work around PI3K inhibition—the very thing we would want to avoid, under this theory.

This situation came up at a dinner in 2014, when I joined Cantley, who was then director of the Meyer Cancer Center at Weill Cornell Medical College in Manhattan, and Siddhartha Mukherjee, who is a practicing oncologist, research scientist, and Pulitzer Prize–winning author of The Emperor of All Maladies, a “biography” of cancer. I was a huge fan of Sid’s work, so I was excited to sit down with these two giants in oncology.

Over dinner, I shared a story about a case where a PI3K-inhibiting drug treatment had been enhanced by a kind of metabolic therapy. I couldn’t stop thinking about it, because the patient was the wife of a very close friend of mine. Sandra (not her real name) had been diagnosed with breast cancer six years earlier. It had already spread to her lymph nodes and her bones. Because of her poor prognosis, she qualified for a clinical trial of an experimental PI3K-inhibitor drug, in combination with standard therapies.

Sandra was a very motivated patient. From the day of her diagnosis, she had become obsessed with doing anything possible to stack the odds in her favor. She devoured everything she could read on the impact of nutrition on cancer, and she had concluded that a diet that reduced insulin and IGF-1 would aid in her treatment. So she worked out a regimen that consisted primarily of leafy vegetables, olive oil, avocados, nuts, and modest amounts of protein, mostly from fish, eggs, and poultry. The diet was just as notable for what it did not contain: added sugar and refined carbohydrates. All along, she underwent frequent blood tests to make sure her insulin and IGF-1 levels stayed low, which they did.

Over the next few years, every other woman who was enrolled at her trial site had died. Every single one. The patients had been on state-of-the-art chemotherapy plus the PI3K inhibitor, yet their metastatic breast cancer had still overtaken them. The trial had to be stopped because it was clear that the drugs were not working. Except for Sandra. Why was she still alive, while hundreds of other women with the same disease, at the same stage, were not? Was she merely lucky? Or could her very strict diet, which likely inhibited her insulin and IGF-1, have played a role in her fate?

I had a hunch that it may have. I believe that we have to pay attention to these outliers, these “miraculous” survivors. Even if they are only anecdotal, their stories may contain some useful insight into this lethal, mysterious disease. As Steve Rosenberg used to say, “These patients help us ask the right questions.”

Yet in his 592-page magnum opus on cancer, published in 2010, Mukherjee had barely written a word about metabolism and metabolic therapies. It seemed premature to write about it, he later told me. Now, as I told this story over dinner, he seemed interested but skeptical. Cantley grabbed a napkin and started scribbling a graphic: the problem with PI3K inhibitors, he explained, is that by turning down the insulin-related PI3K pathway, they actually end up raising insulin and glucose levels. Because glucose is blocked from entering the cell, more of it stays in the bloodstream. The body then thinks it needs to produce more insulin to get rid of all that glucose, possibly negating some of the effects of the drug by activating PI3K. So what if we combined PI3K inhibitors with an insulin-minimizing or ketogenic diet?

From that crude drawing on a napkin, a study was born. Published in Nature in 2018, with Mukherjee and Cantley as senior authors, the study found that a combination of a ketogenic diet and PI3K inhibitors improved the responses to treatment of mice that had been implanted with human cancer tumors. The results are important because they show not only that a cancer cell’s metabolism is a valid target for therapy but that a patient’s metabolic state can affect the efficacy of a drug. In this case, the animals’ ketogenic diet seemed to synergize with what was otherwise a somewhat disappointing treatment, and together they proved to be far more powerful than either one alone. It’s like in boxing, where a combination often proves to be much more effective than any single punch. If your first punch misses, the second is already in motion, aimed directly at the spot where you anticipate your opponent will move. (Mukherjee and Cantley have since partnered on a start-up company to further explore this idea of combining drug treatment with nutritional interventions.)

Other types of dietary interventions have been found to help improve the effectiveness of chemotherapy, while limiting its collateral damage to healthy tissues. Work by Valter Longo of the University of Southern California and others has found that fasting, or a fasting-like diet, increases the ability of normal cells to resist chemotherapy, while rendering cancer cells more vulnerable to the treatment. It may seem counterintuitive to recommend fasting to cancer patients, but researchers have found that it caused no major adverse events in chemotherapy patients, and in some cases it may have improved the patients’ quality of life. A randomized trial in 131 cancer patients undergoing chemotherapy found that those who were placed on a “fasting-mimicking diet” (basically, a very low-calorie diet designed to provide essential nutrients while reducing feelings of hunger) were more likely to respond to chemotherapy and to feel better physically and emotionally.

This flies in the face of traditional practice, which is to try to get patients on chemotherapy to eat as much as they can tolerate, typically in the form of high-calorie and even high-sugar diets. The American Cancer Society suggests using ice cream “as a topping on cake.” But the results of these studies suggest that maybe it’s not such a good idea to increase the level of insulin in someone who has cancer. More studies need to be done, but the working hypothesis is that because cancer cells are so metabolically greedy, they are therefore more vulnerable than normal cells to a reduction in nutrients—or more likely, a reduction in insulin, which activates the PI3K pathway essential to the Warburg effect.

This study and the Mukherjee-Cantley study we discussed earlier also point toward another important takeaway from this chapter, which is that there is rarely only one way to treat a cancer successfully. As Keith Flaherty, a medical oncologist and the director of developmental therapeutics at Massachusetts General Hospital, explained to me, the best strategy to target cancer is likely by targeting multiple vulnerabilities of the disease at one time, or in sequence. By stacking different therapies, such as combining a PI3K inhibitor with a ketogenic diet, we can attack cancer on multiple fronts, while also minimizing the likelihood of the cancer developing resistance (via mutations) to any single treatment. This is becoming a more common practice in conventional chemotherapy—but for it to be truly effective, we need more efficacious treatments to work with in the first place, ones that do a better job of singling out cancer cells for destruction while leaving healthy cells, and the patient, unharmed.

In the next section, we’ll look at how the once-outlandish notion of immunotherapy has yielded multiple potentially game-changing cancer therapies that could fit this bill.

The Promise of Immunotherapy

Like metabolism, immunotherapy did not figure in The Emperor of All Maladies. It was barely on the radar when the book was published in 2010. But when Ken Burns made his documentary based on the book just five years later, both immunotherapy and Steve Rosenberg were featured very prominently, which speaks to the degree to which our thinking about cancer and especially immunotherapy has begun to change, just in the last decade.

The immune system is programmed to distinguish “nonself” from “self”—that is, to recognize invading pathogens and foreign bodies among our own healthy native cells, and then to kill or neutralize the harmful agents. An immunotherapy is any therapy that tries to boost or harness the patient’s immune system to fight an infection or other condition (example: vaccines). The problem with trying to treat cancer this way is that while cancer cells are abnormal and dangerous, they are technically still our cells (“self”). They have cleverly evolved to hide from the immune system and specifically our T cells, the immune system’s assassins that would ordinarily kill foreign cells. So for a cancer immunotherapy to succeed, we essentially need to teach the immune system to recognize and kill our own cells that have turned cancerous. It needs to be able to distinguish “bad self” (cancer) from “good self” (everything else).

Rosenberg wasn’t the first person to try to harness the immune system against cancer. Back in the late nineteenth century, a Harvard-trained surgeon named William Coley had noticed that a patient with a serious tumor had been miraculously cured, apparently as a result of a major postsurgical infection. Coley began experimenting with bacterial inoculations that he hoped would trigger a similar immune response in other patients. But his medical colleagues were appalled by the notion of injecting patients with germs, and when others failed to reproduce Coley’s results, his ideas were sidelined and condemned as quackery. Yet cases of spontaneous cancer remission—like the one Rosenberg had observed as a young resident—kept occurring, and nobody could really explain them. They offered a tantalizing glimpse of the healing power of the human body.

It wasn’t an easy problem to solve. Rosenberg tried one approach after another, without success. A cancer “vaccine” went nowhere. He spent many years experimenting with interleukin-2 (IL-2), a cytokine that plays a major role in the immune response (it basically amplifies the activity of lymphocytes, white blood cells that fight infection). It had worked in animal models of metastatic cancer, but results in human subjects were more mixed: patients had to spend days, if not weeks, in the ICU, and they could very easily die from its massive side effects. Finally, in 1984, a late-stage melanoma patient named Linda Taylor was put into remission by high-dose IL-2 alone.

This was a huge turning point, because it showed that the immune system could fight off cancer. But the failures still outnumbered the successes, as high-dose IL-2 seemed to be effective only against melanoma and renal cell cancers, and only in 10 to 20 percent of patients with these two cancers.[*2] It was a shotgun approach to a problem that required more precision. So Rosenberg turned his attention to the T cells directly. How could they be trained to spot and attack cancer cells?

It took many more years and several iterations, but Rosenberg and his team adapted a technique that had been developed in Israel that involved taking T cells from a patient’s blood, then using genetic engineering to add antigen receptors that were specifically targeted to the patient’s tumors. Now the T cells were programmed to attack the patient’s cancer. Known as chimeric antigen receptor T cells (or CAR-T), these modified T cells could be multiplied in the lab and then infused back into the patient.

In 2010, Rosenberg and his team reported their first success with a CAR-T treatment, in a patient with advanced follicular lymphoma who had undergone multiple rounds of conventional treatments, including chemotherapy, as well as a different kind of immunotherapy, all without success. Other groups pursued the technique as well, and finally, in 2017, the first two CAR-T-based treatments were approved by the FDA (making them the first cell and gene therapies ever approved by the FDA), one for adult lymphoma and another for acute lymphoblastic leukemia, the most common cancer in children. It had taken nearly fifty years, but Steve Rosenberg’s once-outlandish theory had finally yielded a breakthrough.


As elegant as they are, however, CAR-T treatments have proven successful only against one specific type of cancer called B-cell lymphoma. All B-cells, normal and cancerous alike, express a protein called CD19, which is the target used by the CAR-T cell to zero in and kill them. Since we can live without B-cells, CAR-T works by obliterating all CD19-bearing cells. Unfortunately, we have not yet identified a similar marker for other cancers.

If we are going to bring down overall cancer death rates, we need a more broadly successful class of treatments. Luckily, the immunotherapy approach continued to evolve. Now, a bit more than a decade later, a handful of immunotherapy-based cancer drugs have been approved. In addition to CAR-T, there is a class of drugs called “checkpoint inhibitors,” which take an opposite approach to the T cell–based therapies. Instead of activating T cells to go kill the cancer, the checkpoint inhibitors help make the cancer visible to the immune system.

To simplify a very long and fascinating story,[*3] a researcher from Texas named James Allison, who has been working on immunotherapy for almost as long as Steve Rosenberg, figured out how cancer cells hide from the immune system by exploiting so-called checkpoints that are normally supposed to regulate our T cells and keep them from going overboard and attacking our normal cells, which would lead to autoimmune disease. Essentially, the checkpoints ask the T cells, one last time, “Are you sure you want to kill this cell?”

Allison found that if you blocked specific checkpoints, particularly one called CTLA-4, you effectively outed or unmasked the cancer cells, and the T cells would then destroy them. He tried the technique on cancer-prone mice, and in one early experiment he arrived at his lab one morning to find that all the mice who had received checkpoint-inhibiting therapy were still alive, while the ones without it were all dead. It’s nice when your results are so clear that you don’t even really need statistical analysis.

In 2018, Allison shared the Nobel Prize with a Japanese scientist named Tasuku Honjo, who had been working on a slightly different checkpoint called PD-1. The work of these two scientists has led to two approved checkpoint-inhibiting drugs, ipilimumab (Yervoy) and pembrolizumab (Keytruda), targeting CTLA-4 and PD-1, respectively.

All Nobel Prizes are impressive, but I’m pretty biased toward this one. Checkpoint inhibitors not only saved the life of a third Nobel laureate, former US president Jimmy Carter, who was treated with Keytruda for his metastatic melanoma in 2015, but they also came to the rescue of a very good friend of mine, a former colleague whom I’ll call Michael. When he was only in his early forties, Michael was diagnosed with a very large colon tumor that required immediate surgery. I still remember the cover of the magazine that I flipped through anxiously while I was sitting in the waiting room during his procedure. Michael is the kindest soul I think I’ve ever known, and his brilliance and wit could make the worst day seem enjoyable during the years we worked together. I could not imagine losing him.

His family and friends were elated when his surgery proved successful and the pathology report found no sign of cancer in his nearby lymph nodes, despite the advanced size of the primary tumor. A few months later, our joy was turned back to despair as we learned that Michael’s cancer had been the result of a genetic condition called Lynch syndrome. People with Lynch syndrome usually know they have it, because it is inherited in a dominant fashion. But Michael had been adopted, so he had no idea he was at risk. The mutations that define Lynch syndrome all but guarantee that its carriers develop early-onset colon cancer, as Michael had, but they are also at very high risk for other cancers. Sure enough, five years after he dodged that first bullet with colon cancer, Michael called to say that he now had pancreatic adenocarcinoma. This was even more distressing, because as we both well knew, this cancer is almost uniformly fatal.

Michael went to see the top pancreatic surgeon in his area, who confirmed the worst: surgery was not possible. The cancer was too advanced. Michael would have, at most, nine to twelve months to live. Making this all the more heart-wrenching was the fact that Michael and his wife had just welcomed their first children, twin girls, into the world that year. But The New England Journal of Medicine had recently reported that some patients with mismatch-repair deficiency (common in Lynch syndrome) had been successfully treated with Keytruda, the anti-PD-1 drug. It was a long shot, but it seemed at least plausible that Michael might benefit from this drug. Michael’s doctors agreed to test him, and the tests confirmed that Michael was indeed a candidate for Keytruda. He was immediately enrolled in a clinical trial. While it is not guaranteed to work on all such patients, it worked on Michael, turning his immune system against his tumor and eventually eradicating all signs of pancreatic cancer in his body.

So now he is cancer-free for a second time, and beyond grateful to have survived a disease that should have killed him when his twin girls were still in diapers. Now he gets to see them grow up. The price he paid is that while his immune system was attacking his cancer, it went a bit overboard and also destroyed his pancreas in the process. As a result, he now has type 1 diabetes, since he can’t produce insulin anymore. He lost his pancreas, but his life was saved. Seems like a fair trade, on balance.


Michael was one of the lucky ones. As of yet, the various immunotherapy treatments that have been approved still benefit only a fairly small percentage of patients. About a third of cancers can be treated with immunotherapy, and of those patients, just one-quarter will actually benefit (i.e., survive). That means that only 8 percent of potential cancer deaths could be prevented by immunotherapy, according to an analysis by oncologists Nathan Gay and Vinay Prasad. They work when they work, but only in a small number of patients and at great cost. Nevertheless, just two decades ago—when I was feeling frustration as a cancer surgeon in training—patients like former president Carter or my friend Michael, and countless others, would have all died.

But I (and others who know much more than I do) believe that we have had only a small taste of what can be accomplished via immunotherapy.

One idea that is currently being explored is the notion of combining immunotherapies with other treatments. One recent paper describes a clinical trial where a platinum-based chemotherapy was used in combination with a checkpoint inhibitor, resulting in improved overall survival in patients with lung cancer. These patients were not sensitive to the checkpoint inhibitor alone, but something about the chemo made the cancer more sensitive, or more “visible” if you will, to the immunotherapy. It’s an extension of the idea of “stacking” therapies that we mentioned earlier.

To make immunotherapy more widely effective, we need to devise ways to help our immune cells detect and kill a broader array of cancers, not just a few specific types. Genetic analysis reveals that some 80 percent of epithelial cancers (that is, solid organ tumors) possess mutations that the immune system can recognize—thus making them potentially vulnerable to immune-based treatments.


One very promising technique is called adoptive cell therapy (or adoptive cell transfer, ACT). ACT is a class of immunotherapy whereby supplemental T cells are transferred into a patient, like adding reinforcements to an army, to bolster their ability to fight their own tumor. These T cells have been genetically programmed with antigens specifically targeted at the patient’s individual tumor type. It is similar to CAR-T cell therapy, which we discussed earlier, but much broader in scope. As cancer grows, it quickly outruns the immune system’s ability to detect and kill it; there simply aren’t enough T cells to do the job, particularly when the cancer reaches the point of clinical detection. This is why spontaneous remission, as happened with James DeAngelo, is so rare. The idea behind ACT is basically to overwhelm the cancer with a huge number of targeted T cells, like supplementing an army with a brigade of trained assassins.

There are two ways to do ACT. First, we can take a sample of a patient’s tumor and isolate those T cells that do recognize the tumor as a threat. These are called tumor-infiltrating lymphocytes (TILs), but there may only be a few million of them, not enough to mount a complete response against the tumor. By removing the TILs from the body and multiplying them by a factor of 1,000 or so, and then reinfusing them into the patient, we can expect to see a much better response. Alternatively, T cells can be harvested from the patient’s blood and genetically modified to recognize his or her specific tumor. Each of these approaches has advantages and disadvantages,[*4] but the interesting part is that ACT effectively means designing a new, customized anticancer drug for each individual patient.

This is obviously a costly proposition, and a very labor-intensive process, but it has a lot of promise. The proof of principle is here, but much more work is needed, not only to improve the efficacy of this approach but also to enable us to deliver this treatment more widely and more easily. And while the cost may seem prohibitive at first, I would point out that conventional chemotherapy is also very expensive—and its remissions are almost never permanent.

One striking feature of immune-based cancer treatment is that when it works, it really works. It is not uncommon for a patient with metastatic cancer to enter remission after chemotherapy. The problem is that it virtually never lasts. The cancer almost always comes back in some form. But when patients do respond to immunotherapy, and go into complete remission, they often stay in remission. Between 80 and 90 percent of so-called complete responders to immunotherapy remain disease-free fifteen years out. This is extraordinary—far better than the short-term, five-year time horizon at which we typically declare victory in conventional cancer treatment. One hesitates to use the word cured, but in patients who do respond to immunotherapy, it’s safe to assume that the cancer is pretty much gone.

The important message here is that there is hope. For the first time in my lifetime, we are making progress in the War on Cancer, if we can even still call it that. There are now treatments that can, and do, save the lives of thousands of people who would have inevitably died just a decade ago. Twenty years ago, someone with metastatic melanoma could expect to live about six more months, on average. Now that number is twenty-four months, with about 20 percent of such patients being completely cured. This represents measurable progress—almost entirely thanks to immunotherapy. Improved early detection of cancer, which we will discuss in the final section of this chapter, will likely make our immunotherapy treatments still more effective.

Immunotherapy traveled a rocky road, and it could have been abandoned completely at many points along the way. In the end, it survived because its chimerical successes turned out to be not so chimerical, but mostly thanks to the determination and persistence of visionary scientists such as James Allison, Tasuku Honjo, Steve Rosenberg, and others, who kept going even when their work seemed pointless and possibly crazy.

Early Detection

The final and perhaps most important tool in our anticancer arsenal is early, aggressive screening. This remains a controversial topic, but the evidence is overwhelming that catching cancer early is almost always net beneficial.

Unfortunately, the same problem that I encountered in residency applies today: too many cancers are detected too late, after they’ve grown and spread via metastasis. Very few treatments work against these advanced cancers; in most cases, outside of the few cancers that respond to immunotherapies, the best we can hope for is to delay death slightly. The ten-year survival rate for patients with metastatic cancer is virtually the same now as it was fifty years ago: zero. We need to do more than hope for novel therapies.

When cancers are detected early, in stage I, survival rates skyrocket. This is partly because of simple math: these earlier-stage cancers comprise fewer total cancerous cells, with fewer mutations, and thus are more vulnerable to treatment with the drugs that we do have, including some immunotherapies. I would go so far as to argue that early detection is our best hope for radically reducing cancer mortality.

This claim makes intuitive sense, but it is supported by even a cursory look at data comparing success rates at treating specific cancers in the metastatic versus adjuvant (i.e., postsurgical) setting. Let’s look at colon cancer first. A patient with metastatic colon cancer, which means the cancer has spread past their colon and adjacent lymph nodes to another part of the body, such as the liver, will typically be treated with a combination of three drugs known as the FOLFOX regimen. This treatment yields a median survival time of about 31.5 months,[*5] meaning about half of patients live longer than this, and half do not. Regardless, virtually none of these patients will be alive in ten years. If a patient undergoes successful surgery for stage III colon cancer, which means all the cancer was removed and there was no visible spread to distant organs, then the follow-up care is treatment with the exact same FOLFOX treatment regimen. But in this scenario, fully 78.5 percent of these patients will survive for another six years—more than twice as long as median survival for the metastatic patients—and 67 percent of them will still be alive ten years after surgery. That’s an impressive difference.

So what explains it? The difference has to do with the overall burden of cancer cells in each patient. In advanced, metastatic cancer there are tens if not hundreds of billions of cancer cells in need of treatment. In less advanced cancers, while there are undoubtedly still millions, if not billions, of cancer cells that have escaped the surgeon’s scalpel, the far lower population means they will also have fewer mutations and thus less resistance to the treatment.

It’s a similar story for patients with breast cancer. Patients with HER2-positive metastatic breast cancer[*6] can expect a median survival time of just under five years, with standard treatment consisting of three chemotherapy drugs. But if our patient has a smaller (< 3 cm), localized, HER2+ tumor that is removed surgically, plus adjuvant treatment with just two of these chemo drugs, she will have a 93 percent chance of living for at least another seven years without disease. The lower a patient’s overall tumor burden, the more effective our drugs tend to be—and the greater the patient’s odds of survival. Again, just as with immunotherapy treatment, it’s a numbers game: the fewer cancerous cells we have, the greater our likelihood of success.

The problem is that we’re still not very good at detecting cancer in these early stages—yet. Out of dozens of different types of cancers, we have agreed-upon, reliable screening methods for only five: lung (for smokers), breast, prostate, colorectal, and cervical. Even so, mainstream guidelines have been waving people away from some types of early screening, such as mammography in women and blood testing for PSA, prostate-specific antigen, in men. In part this has to do with cost, and in part this has to do with the risk of false positives that may lead to unnecessary or even dangerous treatment (entailing further costs). Both are valid issues, but let’s set aside the cost issue and focus on the false-positive problem.

Medicine 2.0 says that because there are significant false positives with certain tests, we shouldn’t do these tests on most people, period. But if we put on our Medicine 3.0 glasses, we see it differently: these tests are potentially useful, and they’re just about all we have. So how can we make them more useful and accurate?

With all diagnostic tests, there is a trade-off between sensitivity, or the ability of the test to detect an existing condition (i.e., its true positive rate, expressed as a percentage), and specificity, which is the ability to determine that someone does not have that condition (i.e., true negative rate). Together, these represent the test’s overall accuracy. In addition, however, we have to consider the prevalence of the disease in our target population. How likely is it that the person we are testing actually has this condition? Mammography has a sensitivity in the mideighties and a specificity in the low nineties. But if we are examining a relatively low-risk population, perhaps 1 percent of whom actually have breast cancer, then even a test with decent sensitivity is going to generate a fairly large number of false positives. In fact, in this low-risk group, the “positive predictive value” of mammography is only about 10 percent—meaning that if you do test positive, there is only about a one-in-ten chance that you actually have breast cancer. In other populations, with greater overall prevalence (and risk), the test performs much better.

The situation with mammography illustrates why we need to be very strategic about who we are testing and what their risk profile might be, and to understand what our test can and can’t tell us. No single diagnostic test, for anything, is 100 percent accurate. So it is foolish to rely on just one test, not only for breast cancer but in many other areas as well. We need to think in terms of stacking test modalities—incorporating ultrasound and MRI in addition to mammography, for example, when looking for breast cancer. With multiple tests, our resolution improves and fewer unnecessary procedures will be performed.

In short, the problem is not the tests themselves but how we use them. Prostate cancer screening provides an even better example. It’s no longer as simple as “Your PSA number is X or higher, and therefore we must biopsy your prostate, a painful procedure with many unpleasant possible side effects.” Now we know to look at other parameters, such as PSA velocity (the speed at which PSA has been changing over time), PSA density (PSA value normalized to the volume of the prostate gland), and free PSA (comparing the amount of PSA that is bound versus unbound to carrier proteins in the blood). When those factors are taken into account, PSA becomes a much better indicator of prostate cancer risk.

Then there are other tests, such as the 4K blood test, which looks for specific proteins that might give us a better idea of how aggressive and potentially dangerous the patient’s prostate cancer might be. The key question we want to answer is, Will our patient die with prostate cancer, as many men do, or will he die from it? We’d rather not disrupt his life, and potentially do him harm, in the course of finding out. Combining these blood tests I’ve just described with the techniques of multiparametric MRI imaging means that the likelihood of performing an unnecessary biopsy or surgery is now very low.

There is a similar but quieter controversy around screening for colorectal cancer (CRC), which has long been a rite of passage for those in middle age.[*7] The purpose of the colonoscopy is to look not only for full-fledged tumors but also for polyps, which are growths that form in the lining of the colon. Most polyps remain small and harmless and never become cancerous, but some have the potential to become malignant and invade the wall of the colon. Not all polyps become cancer, but all colon cancers came from polyps. This is what makes a colonoscopy such a powerful tool. The endoscopist is able not only to spot potentially cancerous growths before they become dangerous but also to intervene on the spot, using instruments on the colonoscope to remove polyps for later examination. It combines screening and surgery into one procedure. It’s an amazing tool.

Traditional guidelines have recommended colorectal cancer screenings for average-risk people ages fifty to seventy-five. These preventive screenings are fully covered under the Affordable Care Act, and if no polyps are found and the patient is of average risk, the procedure needs to be repeated only every ten years, according to consensus guidelines. But there is ample evidence out there that age fifty may be too old for a first screening, even in patients with average risk factors (that is, no family history of colon cancer and no personal history of inflammatory bowel disease). About 70 percent of people who are diagnosed with CRC before the age of fifty have no family history or hereditary conditions linked to the disease. In 2020, some 3,640 Americans died from colorectal cancer before they turned fifty—and given the slow-moving nature of the disease, it’s likely that many of those who died later than that already had the disease on their fiftieth birthday. This is why the American Cancer Society updated its guidelines in 2018, lowering the age to forty-five for people at average risk.

In my practice, we go further, typically encouraging average-risk individuals to get a colonoscopy by age forty—and even sooner if anything in their history suggests they may be at higher risk. We then repeat the procedure as often as every two to three years, depending on the findings from the previous colonoscopy. If a sessile (flat) polyp is found, for example, we’re inclined to do it sooner than if the endoscopist finds nothing at all. Two or three years might seem like a very short window of time to repeat such an involved procedure, but colon cancer has been documented to appear within the span of as little as six months to two years after a normal colonoscopy. Better safe than sorry.[*8]

Why do I generally recommend a colonoscopy before the guidelines do? Mostly because, of all the major cancers, colorectal cancer is one of the easiest to detect, with the greatest payoff in terms of risk reduction. It remains one of the top five deadliest cancers in the United States, behind lung (#1) and breast/prostate (#2 for women/men), and just ahead of pancreas (#4) and liver (#5) cancers. Of these five, though, CRC is the one we have the best shot at catching early. As it grows in a relatively accessible location, the colon, we can see it without any need for imaging techniques or a surgical biopsy. Because it is so easily observed, we understand its progression from normal tissue to polyp to tumor. Finding it early makes a huge difference, since we can effectively eliminate polyps or growths on the spot. If only we could do that with arterial plaques.

My bottom line is that it is far better to screen early than risk doing it too late. Think asymmetric risk: It’s possible that not screening early and frequently enough is the most dangerous option.[*9]

Other cancers that are relatively easy to spot on visual examination include skin cancer and melanomas. The pap smear for cervical cancer is another well-established, minimally invasive test that I recommend my patients do yearly. When we’re talking about cancers that develop inside the body, in our internal organs, things get trickier. We can’t see them directly, so we must rely on imaging technologies such as low-dose CT scans for lung cancer. These scans are currently recommended in smokers and former smokers, but (as always) I think they should be used more widely, because about 15 percent of lung cancers are diagnosed in people who have never smoked. Lung cancer is the #1 cause of cancer deaths overall, but lung cancer in never-smokers ranks seventh, all by itself.

MRI has a distinct advantage over CT in that it does not produce any ionizing radiation but still provides good resolution. One newer technique that can enhance the ability of a screening MRI to differentiate between a cancer and noncancer is something called diffusion-weighted imaging with background subtraction, or DWI for short. The idea behind DWI is to look at water movement in and around tissue, at different points in time very close to each other (between ten and fifty microseconds, typically). If the water is held or trapped, then it could indicate the presence of a tightly packed cluster of cells, a possible tumor. So the higher the density of cells, the brighter the signal on the DWI phase of MRI, making DWI functionally a radiographic “lump detector.” Right now, DWI works best in the brain because it suffers least from movement artifacts.

I remain optimistic that this technique can be improved over time, with optimization of software and standardization of technique. Despite all this, even something as advanced as the best DWI MRI is not without problems, if used in isolation. While the sensitivity of this test is very high (meaning it’s very good at finding cancer if cancer is there, hence very few false negatives), the specificity is relatively low (which means it’s not as good at telling you when you don’t have cancer, hence a lot of false positives). This is the inevitable trade-off, the yin and yang if you will, between sensitivity and specificity. The more you increase one, the more you decrease the other.[*10]

I tell patients, if you’re going to have a whole-body screening MRI, there is a good chance we’ll be chasing down an insignificant thyroid (or other) nodule in exchange for getting such a good look at your other organs. As a result of this, about a quarter of my patients, understandably, elect not to undergo such screening. Which brings me to the next tool in the cancer screening tool kit, a tool that can complement the high sensitivity / low specificity problem of imaging tests.

I am cautiously optimistic about the emergence of so-called “liquid biopsies” that seek to detect the presence of cancers via a blood test.[*11] These are used in two settings: to detect recurrences of cancer in patients following treatment and to screen for cancers in otherwise healthy patients, a fast-moving and exciting field called multicancer early detection.

Max Diehn, one of my med school classmates and now an oncology professor at Stanford, has been on the forefront of this research since 2012. Max and his colleagues set out to ask a seemingly simple question initially. After a patient with lung cancer has had a resection of their tumor, is there any way a blood test can be used to screen the patient for signs of tumor recurrence?

Historically, this has been done via imaging tests, such as CT scans, that enable us to “see” a tumor. Radiation exposure notwithstanding, the main issue is that these tests don’t have very high resolution. It’s very difficult for these imaging technologies to discern a cancer smaller than about one centimeter in diameter. Even if you assume that this one-centimeter nodule is the only collection of cancer cells in the patient’s body (not a great assumption; the mouse in the trap is rarely the only one in the house), you’re still talking about more than a billion cancer cells by the time you reach the threshold of traditional detection. If we could catch these recurrent cancers sooner, we might have a better shot at keeping patients in remission—for the same reasons that it’s easier to treat adjuvant versus metastatic cancer, as we discussed a few pages ago.

Max and his colleagues came up with a wholly different method. Because cancer cells are growing constantly, they tend to shed cellular matter, including bits of tumor DNA, into the circulation. What if there were a blood test that could detect this so-called cell-free DNA? We would already know the genetic signature of the tumor from the surgery—that is, how the lung cancer cells differ from normal lung cells. Thus, it should be possible to screen for this cell-free DNA in a patient’s plasma and thereby determine the presence of cancer.

Make no mistake about it, this is still akin to looking for a needle in a haystack. In an early-stage cancer, which are the cancers we would most want to find via liquid biopsies, we might be talking about 0.01 to 0.001 percent of cell-free DNA coming from the cancer (or about one part in ten thousand to one hundred thousand). Only with the help of next-generation, high-throughput DNA screening technology is this possible. These tests are becoming more widely used in the postsurgical setting, but the technology is still relatively young. The key is that you must know what you’re looking for—the patterns of mutations that distinguish cancer from normal cells.

Some researchers are beginning to develop ways to use blood tests to screen for cancer generally, in otherwise healthy people. This is an order of magnitude more difficult, like looking for a needle in ten haystacks; worse, in this case we don’t even know what the needle should look like. We don’t know anything about the patient’s tumor mutation patterns, because we’re not yet certain that they have cancer. Thus we must look for other potential markers. One company leading the charge with this type of assay is called Grail, a subsidiary of the genetic-sequencing company Illumina. The Grail test, known as Galleri, looks at methylation patterns of the cell-free DNA, which are basically chemical changes to the DNA molecules that suggest the presence of cancer. Using very-high-throughput screening and a massive AI engine, the Galleri test can glean two crucial pieces of information from this sample of blood: Is cancer present? And if so, where is it? From what part of the body did it most likely originate?

With any diagnostic test, a decision has to be made with respect to how to calibrate, or tune, it. Is it going to be geared toward higher sensitivity or higher specificity? Galleri has been validated against a database called the Circulating Cell-free Genome Atlas (CCGA), which is based on blood samples from more than fifteen thousand patients both with and without cancer. In this study, the Galleri test proved to have a very high specificity, about 99.5 percent, meaning only 0.5 percent of tests yielded a false positive. If the test says you have cancer, somewhere in your body, then it is likely that you do. The trade-off is that the resulting sensitivity can be low, depending on the stage. (That is, even if the test says you don’t have cancer, you are not necessarily in the clear.)

The thing to keep in mind here, however, is that this test still has much higher resolution than radiographic tests such as MRI or mammogram. Those imaging-based tests require “seeing” the tumor, which can happen only when the tumor reaches a certain size. With Galleri, the test is looking at cell-free DNA, which can come from any size tumor—even ones that remain invisible to imaging tests.

One early observation of the CCGA study was that detectability was associated not only with the stage of tumor, which would be expected (the more advanced the tumor, the greater the likelihood of finding cell-free DNA in the blood), but also with the subtype of tumor. For example, the detection rate for stage I/II hormone receptor–positive breast cancer is about 25 percent, while the detection rate for stage I/II hormone receptor–negative breast cancer is about 75 percent. What does this difference tell us? We know that breast cancer is not a uniform disease and that the hormone receptor–negative tumors are more lethal than hormone receptor–positive tumors. Thus, the test proves more accurate in detecting the more lethal subtype of breast cancer.

Liquid biopsies could be viewed as having two functions: first, to determine cancer’s presence or absence, a binary question; and second and perhaps more important, to gain insight into the specific cancer’s biology. How dangerous is this particular cancer likely to be? The cancers that shed more cell-free DNA, it seems, also tend to be more aggressive and deadly—and are thus the cancers that we want to detect, and treat, as soon as possible. This technology is still in its infancy, but I’m hopeful that pairing different diagnostic tests ranging from radiographic (e.g., MRI) to direct visualization (e.g., colonoscopy) to biological/genetic (e.g., liquid biopsy) will allow us to correctly identify the cancers that need treatment the soonest, with the fewest possible false positives.

The implications of this, I think, are seismic: if liquid biopsies deliver on their promise, we could completely flip the time line of cancer so that we are routinely intervening early, when we have a chance of controlling or even eliminating the cancer—rather than the way we typically do it now, coming in at a late stage, when the odds are already stacked against the patient, and hoping for a miracle.


Of all the Horsemen, cancer is probably the hardest to prevent. It is probably also the one where bad luck in various forms plays the greatest role, such as in the form of accumulated somatic mutations. The only modifiable risks that really stand out in the data are smoking, insulin resistance, and obesity (all to be avoided)—and maybe pollution (air, water, etc.), but the data here are less clear.

We do have some treatment options for cancer, unlike with Alzheimer’s disease (as we’ll see in the next chapter), and immunotherapy in particular has great promise. Yet our treatment and prevention strategies remain far less effective than the tools that we have to address cardiovascular disease and the spectrum of metabolic dysfunction from insulin resistance to type 2 diabetes.

Until we learn how to prevent or “cure” cancer entirely, something I do not see happening in our lifetime, short of some miraculous breakthroughs, we need to focus far more energy on early detection of cancer, to enable better targeting of specific treatments at specific cancers while they are at their most vulnerable stages. If the first rule of cancer is “Don’t get cancer,” the second rule is “Catch it as soon as possible.”

This is why I’m such an advocate for early screening. It’s a simple truth that treating smaller tumors with fewer mutations is far easier than if we wait for the cancer to advance and potentially acquire mutations that help it evade our treatments. The only way to catch it early is with aggressive screening.

This does come at significant cost, which is why Medicine 2.0 tends to be more conservative about screening. There is a financial cost, of course, but there is also an emotional cost, particularly of tests that may generate false positives. And there are other, incidental risks, such as the slight risk from a colonoscopy or the more significant risk from an unnecessary biopsy. These three costs must be weighed against the cost of missing a cancer, or not spotting it early, when it is still susceptible to treatment.

Nobody said this was going to be easy. We still have a very long way to go. But there is finally hope, on multiple fronts—far more than when I was training to become a cancer surgeon. More than fifty years into the War on Cancer, we can finally see a path to a world where a cancer diagnosis typically means an early detection of a treatable problem rather than a late discovery of a grim one. Thanks to better screening and more effective treatments such as immunotherapy, cancer could someday become a manageable disease, perhaps no longer even qualifying as a Horseman.

Skip Notes

*1 This is not the only explanation for how the Warburg effect benefits a cancer cell. Another theory is that it helps protect the tumor from immune cells by making the tumor microenvironment less hospitable because of lower pH (i.e., more acidic) caused by the generation of lactic acid and reactive oxygen species. For an excellent review of these topics, see Liberti and Locasale (2016).

*2 At the time it was not clear why this was the case, but today it’s clear that this approach worked because these two cancers tend to have a large number of genetic mutations, meaning the immune system is more likely to recognize the cancerous cells as harmful and target them.

*3 To learn more about the story of immunotherapy, read Charles Graeber’s 2018 book, Breakthrough, which goes into greater detail about Jim Allison’s work with checkpoint inhibitors.

*4 For example, TILs by definition have already demonstrated an affinity for the tumor; however, they may “age” more as they are multiplied (cells “age” every time they divide), losing some of their potency. Conversely, genetically modified T cells tend to be younger and easier to grow, but they don’t necessarily have the same ability to kill tumors as TILs.

*5 For 95 percent of metastatic colon cancer patients.

*6 This means the expression of human epidermal growth factor receptor 2, which is a protein receptor on the surface of breast cancer cells that promotes growth. This is overexpressed in roughly 30 percent of breast cancers.

*7 There are several different colon cancer screening methods that break down into two categories: stool-based tests and direct visualization tests. Stool-based tests are essentially a screening test for a screening test. A positive stool-based test prompts a direct visualization test of the colon: either a flexible sigmoidoscopy, which allows the endoscopist to view the lower part of the colon (including the sigmoid and descending colon), or a traditional colonoscopy, which examines the entire colon. In my opinion, none of the other tests compares to a colonoscopy.

*8 A study published in 2022 found that the risk of CRC was only reduced by 18 percent (relative) and 0.22 percent (absolute) in people advised to get one colonoscopy in a ten-year period versus those not advised to. However, only 42 percent of those advised to get a colonoscopy actually did so, and they only underwent one during the period of the study. I would argue this was not a test of the efficacy of frequent colonoscopy for prevention of CRC and was, instead, a test of the effectiveness of telling people to get (infrequent) colonoscopy.

*9 For those seeking more detailed guidance, this is what I wrote (Attia 2020a) in a blog post on CRC screening a few years ago: “Before you get your first colonoscopy, there are [a] few things you can do that may improve your risk-to-benefit ratio. You should ask what your endoscopist’s adenoma detection rate (ADR) is. The ADR is the proportion of individuals undergoing a colonoscopy who have one or more adenomas (or colon polyps) detected. The benchmarks for ADR are greater than 30% in men and greater than 20% in women. You should also ask your endoscopist how many perforations he or she has caused, specifically, as well as any other serious complications, like major intestinal bleeding episodes (in a routine screening setting). Another question you should ask is what is your endoscopist’s withdrawal time, defined as the amount of time spent viewing as the colonoscope is withdrawn during a colonoscopy. A longer withdrawal time suggests a more thorough inspection. A 6-minute withdrawal time is currently the standard of care.”

*10 The specificity of MRI is particularly reduced by glandular tissue. MRI is so good at detecting glandular cancer that it significantly overdoes it. The thyroid gland might be the worst offender.

*11 These are called liquid biopsies to distinguish them from traditional solid-tissue biopsies.