The Emperor of All Maladies Read online

Page 5


  Sandler arrived at Children’s Hospital just a few weeks after Farber had received his first package from Lederle. On September 6, 1947, Farber began to inject Sandler with pteroylaspartic acid or PAA, the first of Lederle’s antifolates. (Consent to run a clinical trial for a drug—even a toxic drug—was not typically required. Parents were occasionally cursorily informed about the trial; children were almost never informed or consulted. The Nuremberg code for human experimentation, requiring explicit voluntary consent from patients, was drafted on August 9, 1947, less than a month before the PAA trial. It is doubtful that Farber in Boston had even heard of any such required consent code.)

  PAA had little effect. Over the next month Sandler turned increasingly lethargic. He developed a limp, the result of leukemia pressing down on his spinal cord. Joint aches appeared, and violent, migrating pains. Then the leukemia burst through one of the bones in his thigh, causing a fracture and unleashing a blindingly intense, indescribable pain. By December, the case seemed hopeless. The tip of Sandler’s spleen, more dense than ever with leukemia cells, dropped down to his pelvis. He was withdrawn, listless, swollen, and pale, on the verge of death.

  On December 28, however, Farber received a new version of antifolate from Subbarao and Kiltie, aminopterin, a chemical with a small change from the structure of PAA. Farber snatched the drug as soon as it arrived and began to inject the boy with it, hoping, at best, for a minor reprieve in his cancer.

  The response was marked. The white cell count, which had been climbing astronomically—ten thousand in September, twenty thousand in November, and nearly seventy thousand in December—suddenly stopped rising and hovered at a plateau. Then, even more remarkably, the count actually started to drop, the leukemic blasts gradually flickering out in the blood and then all but disappearing. By New Year’s Eve, the count had dropped to nearly one-sixth of its peak value, bottoming out at a nearly normal level. The cancer hadn’t vanished—under the microscope, there were still malignant white cells—but it had temporarily abated, frozen into a hematologic stalemate in the frozen Boston winter.

  On January 13, 1948, Sandler returned to the clinic, walking on his own for the first time in two months. His spleen and liver had shrunk so dramatically that his clothes, Farber noted, had become “loose around the abdomen.” His bleeding had stopped. His appetite turned ravenous, as if he were trying to catch up on six months of lost meals. By February, Farber noted, the child’s alertness, nutrition, and activity were equal to his twin’s. For a brief month or so, Robert Sandler and Elliott Sandler seemed identical again.

  Sandler’s remission—unprecedented in the history of leukemia—set off a flurry of activity for Farber. By the early winter of 1948, more children were at his clinic: a three-year-old boy brought with a sore throat, a two-and-a-half-year-old girl with lumps in her head and neck, all eventually diagnosed with childhood ALL. Deluged with antifolates from Yella and with patients who desperately needed them, Farber recruited additional doctors to help him: a hematologist named Louis Diamond, and a group of assistants, James Wolff, Robert Mercer, and Robert Sylvester.

  Farber had infuriated the authorities at Children’s Hospital with his first clinical trial. With this, the second, he pushed them over the edge. The hospital staff voted to take all the pediatric interns off the leukemia chemotherapy unit (the atmosphere in the leukemia wards, it was felt, was far too desperate and experimental and thus not conducive to medical education)—in essence, leaving Farber and his assistants to perform all the patient care themselves. Children with cancer, as one surgeon noted, were typically “tucked in the farthest recesses of the hospital wards.” They were on their deathbeds anyway, the pediatricians argued; wouldn’t it be kinder and gentler, some insisted, to just “let them die in peace”? When one clinician suggested that Farber’s novel “chemicals” be reserved only as a last resort for leukemic children, Farber, recalling his prior life as a pathologist, shot back, “By that time, the only chemical that you will need will be embalming fluid.”

  Farber outfitted a back room of a ward near the bathrooms into a makeshift clinic. His small staff was housed in various unused spaces in the Department of Pathology—in back rooms, stairwell shafts, and empty offices. Institutional support was minimal. Farber’s assistants sharpened their own bone marrow needles, a practice as antiquated as a surgeon whetting his knives on a wheel. Farber’s staff tracked the disease in patients with meticulous attention to detail: every blood count, every transfusion, every fever, was to be recorded. If leukemia was going to be beaten, Farber wanted every minute of that battle recorded for posterity—even if no one else was willing to watch it happen.

  That winter of 1948, a severe and dismal chill descended on Boston. Snowstorms broke out, bringing Farber’s clinic to a standstill. The narrow asphalt road out to Longwood Avenue was piled with heaps of muddy sleet, and the basement tunnels, poorly heated even in the fall, were now freezing. Daily injections of antifolates became impossible, and Farber’s team backed down to three times a week. In February, when the storms abated, the daily injections started again.

  Meanwhile, news of Farber’s experience with childhood leukemia was beginning to spread, and a slow train of children began to arrive at his clinic. And case by case, an incredible pattern emerged: the antifolates could drive leukemia cell counts down, occasionally even resulting in their complete disappearance—at least for a while. There were other remissions as dramatic as Sandler’s. Two boys treated with aminopterin returned to school. Another child, a two-and-a-half-year-old girl, started to “play and run about” after seven months of lying in bed. The normalcy of blood almost restored a flickering, momentary normalcy to the childhood.

  But there was always the same catch. After a few months of remission, the cancer would inevitably relapse, ultimately flinging aside even the most potent of Yella’s drugs. The cells would return in the bone marrow, then burst out into the blood, and even the most active antifolates would not keep their growth down. Robert Sandler died in 1948, having responded for a few months.

  Yet the remissions, even if temporary, were still genuine remissions—and historic. By April 1948, there was just enough data to put together a preliminary paper for the New England Journal of Medicine. The team had treated sixteen patients. Of the sixteen, ten had responded. And five children—about one-third of the initial group—remained alive four or even six months after their diagnosis. In leukemia, six months of survival was an eternity.

  Farber’s paper, published on June 3, 1948, was seven pages long, jam-packed with tables, figures, microscope photographs, laboratory values, and blood counts. Its language was starched, formal, detached, and scientific. Yet, like all great medical papers, it was a page-turner. And like all good novels, it was timeless: to read it today is to be pitched behind the scenes into the tumultuous life of the Boston clinic, its patients hanging on for life as Farber and his assistants scrambled to find new drugs for a dreadful disease that kept flickering away and returning. It was a plot with a beginning, a middle, and, unfortunately, an end.

  The paper was received, as one scientist recalls, “with skepticism, disbelief, and outrage.” But for Farber, the study carried a tantalizing message: cancer, even in its most aggressive form, had been treated with a medicine, a chemical. In six months between 1947 and 1948, Farber thus saw a door open—briefly, seductively—then close tightly shut again. And through that doorway, he glimpsed an incandescent possibility. The disappearance of an aggressive systemic cancer via a chemical drug was virtually unprecedented in the history of cancer. In the summer of 1948, when one of Farber’s assistants performed a bone marrow biopsy on a leukemic child after treatment with aminopterin, the assistant could not believe the results. “The bone marrow looked so normal,” he wrote, “that one could dream of a cure.”

  And so Farber did dream. He dreamed of malignant cells being killed by specific anticancer drugs, and of normal cells regenerating and reclaiming their physiological spaces; of a whole g
amut of such systemic antagonists to decimate malignant cells; of curing leukemia with chemicals, then applying his experience with chemicals and leukemia to more common cancers. He was throwing down a gauntlet for cancer medicine. It was then up to an entire generation of doctors and scientists to pick it up.

  A Private Plague

  We reveal ourselves in the metaphors we choose for depicting the cosmos in miniature.

  —Stephen Jay Gould

  Thus, for 3,000 years and more, this disease has been known to the medical profession. And for 3,000 years and more, humanity has been knocking at the door of the medical profession for a “cure.”

  —Fortune, March 1937

  Now it is cancer’s turn to be the disease that doesn’t knock before it enters.

  —Susan Sontag, Illness as Metaphor

  We tend to think of cancer as a “modern” illness because its metaphors are so modern. It is a disease of overproduction, of fulminant growth—growth unstoppable, growth tipped into the abyss of no control. Modern biology encourages us to imagine the cell as a molecular machine. Cancer is that machine unable to quench its initial command (to grow) and thus transformed into an indestructible, self-propelled automaton.

  The notion of cancer as an affliction that belongs paradigmatically to the twentieth century is reminiscent, as Susan Sontag argued so powerfully in her book Illness as Metaphor, of another disease once considered emblematic of another era: tuberculosis in the nineteenth century. Both diseases, as Sontag pointedly noted, were similarly “obscene—in the original meaning of that word: ill-omened, abominable, repugnant to the senses.” Both drain vitality; both stretch out the encounter with death; in both cases, dying, even more than death, defines the illness.

  But despite such parallels, tuberculosis belongs to another century. TB (or consumption) was Victorian romanticism brought to its pathological extreme—febrile, unrelenting, breathless, and obsessive. It was a disease of poets: John Keats involuting silently toward death in a small room overlooking the Spanish Steps in Rome, or Byron, an obsessive romantic, who fantasized about dying of the disease to impress his mistresses. “Death and disease are often beautiful, like . . . the hectic glow of consumption,” Thoreau wrote in 1852. In Thomas Mann’s The Magic Mountain, this “hectic glow” releases a feverish creative force in its victims—a clarifying, edifying, cathartic force that, too, appears to be charged with the essence of its era.

  Cancer, in contrast, is riddled with more contemporary images. The cancer cell is a desperate individualist, “in every possible sense, a nonconformist,” as the surgeon-writer Sherwin Nuland wrote. The word metastasis, used to describe the migration of cancer from one site to another, is a curious mix of meta and stasis—“beyond stillness” in Latin—an unmoored, partially unstable state that captures the peculiar instability of modernity. If consumption once killed its victims by pathological evisceration (the tuberculosis bacillus gradually hollows out the lung), then cancer asphyxiates us by filling bodies with too many cells; it is consumption in its alternate meaning—the pathology of excess. Cancer is an expansionist disease; it invades through tissues, sets up colonies in hostile landscapes, seeking “sanctuary” in one organ and then immigrating to another. It lives desperately, inventively, fiercely, territorially, cannily, and defensively—at times, as if teaching us how to survive. To confront cancer is to encounter a parallel species, one perhaps more adapted to survival than even we are.

  This image—of cancer as our desperate, malevolent, contemporary doppelgänger—is so haunting because it is at least partly true. A cancer cell is an astonishing perversion of the normal cell. Cancer is a phenomenally successful invader and colonizer in part because it exploits the very features that make us successful as a species or as an organism.

  Like the normal cell, the cancer cell relies on growth in the most basic, elemental sense: the division of one cell to form two. In normal tissues, this process is exquisitely regulated, such that growth is stimulated by specific signals and arrested by other signals. In cancer, unbridled growth gives rise to generation upon generation of cells. Biologists use the term clone to describe cells that share a common genetic ancestor. Cancer, we now know, is a clonal disease. Nearly every known cancer originates from one ancestral cell that, having acquired the capacity of limitless cell division and survival, gives rise to limitless numbers of descendants—Virchow’s omnis cellula e cellula e cellula repeated ad infinitum.

  But cancer is not simply a clonal disease; it is a clonally evolving disease. If growth occurred without evolution, cancer cells would not be imbued with their potent capacity to invade, survive, and metastasize. Every generation of cancer cells creates a small number of cells that is genetically different from its parents. When a chemotherapeutic drug or the immune system attacks cancer, mutant clones that can resist the attack grow out. The fittest cancer cell survives. This mirthless, relentless cycle of mutation, selection, and overgrowth generates cells that are more and more adapted to survival and growth. In some cases, the mutations speed up the acquisition of other mutations. The genetic instability, like a perfect madness, only provides more impetus to generate mutant clones. Cancer thus exploits the fundamental logic of evolution unlike any other illness. If we, as a species, are the ultimate product of Darwinian selection, then so, too, is this incredible disease that lurks inside us.

  Such metaphorical seductions can carry us away, but they are unavoidable with a subject like cancer. In writing this book, I started off by imagining my project as a “history” of cancer. But it felt, inescapably, as if I were writing not about something but about someone. My subject daily morphed into something that resembled an individual—an enigmatic, if somewhat deranged, image in a mirror. This was not so much a medical history of an illness, but something more personal, more visceral: its biography.

  So to begin again, for every biographer must confront the birth of his subject: Where was cancer “born”? How old is cancer? Who was the first to record it as an illness?

  In 1862, Edwin Smith—an unusual character: part scholar and part huckster, an antique forger and self-made Egyptologist—bought (or, some say, stole) a fifteen-foot-long papyrus from an antiques seller in Luxor in Egypt. The papyrus was in dreadful condition, with crumbling, yellow pages filled with cursive Egyptian script. It is now thought to have been written in the seventeenth century BC, a transcription of a manuscript dating back to 2500 BC. The copier—a plagiarist in a terrible hurry—had made errors as he had scribbled, often noting corrections in red ink in the margins.

  Translated in 1930, the papyrus is now thought to contain the collected teachings of Imhotep, a great Egyptian physician who lived around 2625 BC. Imhotep, among the few nonroyal Egyptians known to us from the Old Kingdom, was a Renaissance man at the center of a sweeping Egyptian renaissance. As a vizier in the court of King Djozer, he dabbled in neurosurgery, tried his hand at architecture, and made early forays into astrology and astronomy. Even the Greeks, encountering the fierce, hot blast of his intellect as they marched through Egypt centuries later, cast him as an ancient magician and fused him to their own medical god, Asclepius.

  But the surprising feature of the Smith papyrus is not magic and religion but the absence of magic and religion. In a world immersed in spells, incantations, and charms, Imhotep wrote about broken bones and dislocated vertebrae with a detached, sterile scientific vocabulary, as if he were writing a modern surgical textbook. The forty-eight cases in the papyrus—fractures of the hand, gaping abscesses of the skin, or shattered skull bones—are treated as medical conditions rather than occult phenomena, each with its own anatomical glossary, diagnosis, summary, and prognosis.

  And it is under these clarifying headlamps of an ancient surgeon that cancer first emerges as a distinct disease. Describing case forty-five, Imhotep advises, “If you examine [a case] having bulging masses on [the] breast and you find that they have spread over his breast; if you place your hand upon [the] breast [and] find them to be cool, the
re being no fever at all therein when your hand feels him; they have no granulations, contain no fluid, give rise to no liquid discharge, yet they feel protuberant to your touch, you should say concerning him: ‘This is a case of bulging masses I have to contend with. . . . Bulging tumors of the breast mean the existence of swellings on the breast, large, spreading, and hard; touching them is like touching a ball of wrappings, or they may be compared to the unripe hemat fruit, which is hard and cool to the touch.’”

  A “bulging mass in the breast”—cool, hard, dense as a hemat fruit, and spreading insidiously under the skin—could hardly be a more vivid description of breast cancer. Every case in the papyrus was followed by a concise discussion of treatments, even if only palliative: milk poured through the ears of neurosurgical patients, poultices for wounds, balms for burns. But with case forty-five, Imhotep fell atypically silent. Under the section titled “Therapy,” he offered only a single sentence: “There is none.”

  With that admission of impotence, cancer virtually disappeared from ancient medical history. Other diseases cycled violently through the globe, leaving behind their cryptic footprints in legends and documents. A furious febrile plague—typhus, perhaps—blazed through the port city of Avaris in 1715 BC, decimating its population. Smallpox erupted volcanically in pockets, leaving its telltale pockmarks on the face of Ramses V in the twelfth century BC. Tuberculosis rose and ebbed through the Indus valley like its seasonal floods. But if cancer existed in the interstices of these massive epidemics, it existed in silence, leaving no easily identifiable trace in the medical literature—or in any other literature.

  More than two millennia pass after Imhotep’s description until we once more hear of cancer. And again, it is an illness cloaked in silence, a private shame. In his sprawling Histories, written around 440 BC, the Greek historian Herodotus records the story of Atossa, the queen of Persia, who was suddenly struck by an unusual illness. Atossa was the daughter of Cyrus, and the wife of Darius, successive Achaemenid emperors of legendary brutality who ruled over a vast stretch of land from Lydia on the Mediterranean Sea to Babylonia on the Persian Gulf. In the middle of her reign, Atossa noticed a bleeding lump in her breast that may have arisen from a particularly malevolent form of breast cancer labeled inflammatory (in inflammatory breast cancer, malignant cells invade the lymph glands of the breast, causing a red, swollen mass).