Читать онлайн книгу «The Emperor of All Maladies» автора Siddhartha Mukherjee

The Emperor of All Maladies
Siddhartha Mukherjee
Winner of the Guardian First Book Award 2011Winner of the Pulitzer Prize for Non-fiction 2011Shortlisted for the Wellcome Trust Book PrizeShortlisted for the Duff Cooper PrizeNow, as cancer becomes an ever more universal experience, the need to understand it, and its treatment, has never been more compelling. In this groundbreaking and award-winning account Siddhartha Mukherjee tells the fascinating story of our relationship with this disease. From brutal early surgical treatments, to Sidney Farber’s hugely risky discovery of chemotherapy, to the author’s treatment of his own patients, he reveals how far we have come in solving one of science's great mysteries and offers a fascinating glimpse of our future progress.


The Emperor of all Maladies

A BIOGRAPHY OF CANCER
SIDDHARTHA MUKHERJEE



Copyright
Fourth Estate
An imprint of HarperCollinsPublishers Ltd. 1 London Bridge Street London SE1 9GF
www.harpercollins.co.uk (http://www.harpercollins.co.uk/)
Copyright © Siddhartha Mukherjee 2011
The right of Siddhartha Mukherjee to be identified as the author of this work has been asserted by him in accordance with the Copyright, Designs and Patents Act 1988
A catalogue record for this book is available from the British Library
All rights reserved under International and Pan-American Copyright Conventions. By payment of the required fees, you have been granted the non-exclusive, non-transferable right to access and read the text of this ebook on-screen. No part of this text may be reproduced, transmitted, down-loaded, decompiled, reverse engineered, or stored in or introduced into any information storage and retrieval system, in any form or by any means, whether electronic or mechanical, now known or hereinafter invented, without the express written permission of HarperCollins ebooks
HarperCollinsPublishers has made every reasonable effort to ensure that any picture content and written content in this ebook has been included or removed in accordance with the contractual and technological constraints in operation at the time of publication
Source ISBN: 9780007250912
Ebook Edition © FEBRUARY 2011 ISBN: 9780007435814
Version: 2017-09-20
To ROBERT SANDLER (1945–1948), and to those who came before and after him.
Illness is the night-side of life, a more onerous citizenship. Everyone who is born holds dual citizenship, in the kingdom of the well and in the kingdom of the sick. Although we all prefer to use only the good passport, sooner or later each of us is obliged, at least for a spell, to identify ourselves as citizens of that other place.
—Susan Sontag

Contents
Cover (#u07df49a8-29eb-5ceb-9ded-d0b9f3cfe633)
Title Page (#uf0f1c3df-7246-5688-90d1-0943dc61c7f7)
Copyright
Dedication (#u39639ffe-1802-50da-9b8d-766952b0c6d1)
Epigraph (#u4488cc49-2cd8-5160-9def-12179e846990)

Prologue

Part One - “Of blacke cholor, without boyling”
“A suppuration of blood”
“A monster more insatiable than the guillotine”
Farber’s Gauntlet
A Private Plague
Onkos
Vanishing Humors
“Remote Sympathy”
A Radical Idea
The Hard Tube and the Weak Light
Dyeing and Dying
Poisoning the Atmosphere
The Goodness of Show Business
The House That Jimmy Built

Part Two - An Impatient War
“They form a society”
“These new friends of chemotherapy”
“The butcher shop”
An Early Victory
Mice and Men
VAMP
An Anatomist’s Tumor
An Army on the March
The Cart and the Horse
“A moon shot for cancer”

Part Three - “Will you turn me out if I can’t get better?”
“In God we trust. All others [must] have data”
“The smiling oncologist”
Knowing the Enemy
Halsted’s Ashes
Counting Cancer

Part Four - Prevention Is the Cure
“Coffins of black”
The Emperor’s Nylon Stockings
“A thief in the night”
“A statement of warning”
Photographic Insert
“Curiouser and curiouser”
“A spider’s web”
STAMP
The Map and the Parachute

Part Five - “A Distorted Version of Our Normal Selves”
“A unitary cause”
Under the Lamps of Viruses
“The hunting of the sarc”
The Wind in the Trees
A Risky Prediction
The Hallmarks of Cancer

Part Six - The Fruits of Long Endeavors
“No one had labored in vain”
New Drugs for Old Cancers
A City of Strings
Drugs, Bodies, and Proof
A Four-Minute Mile
The Red Queen’s Race
Thirteen Mountains

Atossa’s War

Footnotes (#ub1696537-83dd-577a-a567-257f421f5222)
Notes
Glossary
Selected Bibliography
Index
Acknowledgments
Author’s Note
About the Author (#u7fba6f24-3650-5512-b610-920e9c700108)
About the Publisher (#u537bf2d6-c293-5011-98fd-83d4e088bef6)
In 2010, about six hundred thousand Americans, and more than 7 million humans around the world, will die of cancer. In the United States, one in three women and one in two men will develop cancer during their lifetime. A quarter of all American deaths, and about 15 percent of all deaths worldwide, will be attributed to cancer. In some nations, cancer will surpass heart disease to become the most common cause of death.
Prologue (#ulink_fd6f8713-7ece-5d70-8187-baae32627b28)
Diseases desperate grown

By desperate appliance are relieved,
Or not at all.
—William Shakespeare, Hamlet
Cancer begins and ends with people
. In the midst of scientific abstraction, it is sometimes possible to forget this one basic fact. . . . Doctors treat diseases, but they also treat people, and this precondition of their professional existence sometimes pulls them in two directions at once.
—June Goodfield
On the morning of May 19, 2004, Carla Reed, a thirty-year-old kindergarten teacher from Ipswich, Massachusetts, a mother of three young children, woke up in bed with a headache. “Not just any headache,” she would recall later, “but a sort of numbness in my head. The kind of numbness that instantly tells you that something is terribly wrong.”
Something had been terribly wrong for nearly a month. Late in April, Carla had discovered a few bruises on her back. They had suddenly appeared one morning, like strange stigmata, then grown and vanished over the next month, leaving large map-shaped marks on her back. Almost indiscernibly, her gums had begun to turn white. By early May, Carla, a vivacious, energetic woman accustomed to spending hours in the classroom chasing down five- and six-year-olds, could barely walk up a flight of stairs. Some mornings, exhausted and unable to stand up, she crawled down the hallways of her house on all fours to get from one room to another. She slept fitfully for twelve or fourteen hours a day, then woke up feeling so overwhelmingly tired that she needed to haul herself back to the couch again to sleep.
Carla and her husband saw a general physician and a nurse twice during those four weeks, but she returned each time with no tests and without a diagnosis. Ghostly pains appeared and disappeared in her bones. The doctor fumbled about for some explanation. Perhaps it was a migraine, she suggested, and asked Carla to try some aspirin. The aspirin simply worsened the bleeding in Carla’s white gums.
Outgoing, gregarious, and ebullient, Carla was more puzzled than worried about her waxing and waning illness. She had never been seriously ill in her life. The hospital was an abstract place for her; she had never met or consulted a medical specialist, let alone an oncologist. She imagined and concocted various causes to explain her symptoms—overwork, depression, dyspepsia, neuroses, insomnia. But in the end, something visceral arose inside her—a seventh sense—that told Carla something acute and catastrophic was brewing within her body.
On the afternoon of May 19, Carla dropped her three children with a neighbor and drove herself back to the clinic, demanding to have some blood tests. Her doctor ordered a routine test to check her blood counts. As the technician drew a tube of blood from her vein, he looked closely at the blood’s color, obviously intrigued. Watery, pale, and dilute, the liquid that welled out of Carla’s veins hardly resembled blood.
Carla waited the rest of the day without any news. At a fish market the next morning, she received a call.
“We need to draw some blood again,” the nurse from the clinic said.
“When should I come?” Carla asked, planning her hectic day. She remembers looking up at the clock on the wall. A half-pound steak of salmon was warming in her shopping basket, threatening to spoil if she left it out too long.
In the end, commonplace particulars make up Carla’s memories of illness: the clock, the car pool, the children, a tube of pale blood, a missed shower, the fish in the sun, the tightening tone of a voice on the phone. Carla cannot recall much of what the nurse said, only a general sense of urgency. “Come now,” she thinks the nurse said. “Come now.”


I heard about Carla’s case at seven o’clock on the morning of May 21, on a train speeding between Kendall Square and Charles Street in Boston. The sentence that flickered on my beeper had the staccato and deadpan force of a true medical emergency: Carla Reed/New patient with leukemia/14th Floor/Please see as soon as you arrive. As the train shot out of a long, dark tunnel, the glass towers of the Massachusetts General Hospital suddenly loomed into view, and I could see the windows of the fourteenth floor rooms.
Carla, I guessed, was sitting in one of those rooms by herself, terrifyingly alone. Outside the room, a buzz of frantic activity had probably begun. Tubes of blood were shuttling between the ward and the laboratories on the second floor. Nurses were moving about with specimens, interns collecting data for morning reports, alarms beeping, pages being sent out. Somewhere in the depths of the hospital, a microscope was flickering on, with the cells in Carla’s blood coming into focus under its lens.
I can feel relatively certain about all of this because the arrival of a patient with acute leukemia still sends a shiver down the hospital’s spine—all the way from the cancer wards on its upper floors to the clinical laboratories buried deep in the basement. Leukemia is cancer of the white blood cells—cancer in one of its most explosive, violent incarnations. As one nurse on the wards often liked to remind her patients, with this disease “even a paper cut is an emergency.”
For an oncologist in training, too, leukemia represents a special incarnation of cancer. Its pace, its acuity, its breathtaking, inexorable arc of growth forces rapid, often drastic decisions; it is terrifying to experience, terrifying to observe, and terrifying to treat. The body invaded by leukemia is pushed to its brittle physiological limit—every system, heart, lung, blood, working at the knife-edge of its performance. The nurses filled me in on the gaps in the story. Blood tests performed by Carla’s doctor had revealed that her red cell count was critically low, less than a third of normal. Instead of normal white cells, her blood was packed with millions of large, malignant white cells—blasts, in the vocabulary of cancer. Her doctor, having finally stumbled upon the real diagnosis, had sent her to the Massachusetts General Hospital.


In the long, bare hall outside Carla’s room, in the antiseptic gleam of the floor just mopped with diluted bleach, I ran through the list of tests that would be needed on her blood and mentally rehearsed the conversation I would have with her. There was, I noted ruefully, something rehearsed and robotic even about my sympathy. This was the tenth month of my “fellowship” in oncology—a two-year immersive medical program to train cancer specialists—and I felt as if I had gravitated to my lowest point. In those ten indescribably poignant and difficult months, dozens of patients in my care had died. I felt I was slowly becoming inured to the deaths and the desolation—vaccinated against the constant emotional brunt.
There were seven such cancer fellows at this hospital. On paper, we seemed like a formidable force: graduates of five medical schools and four teaching hospitals, sixty-six years of medical and scientific training, and twelve postgraduate degrees among us. But none of those years or degrees could possibly have prepared us for this training program. Medical school, internship, and residency had been physically and emotionally grueling, but the first months of the fellowship flicked away those memories as if all of that had been child’s play, the kindergarten of medical training.
Cancer was an all-consuming presence in our lives. It invaded our imaginations; it occupied our memories; it infiltrated every conversation, every thought. And if we, as physicians, found ourselves immersed in cancer, then our patients found their lives virtually obliterated by the disease. In Aleksandr Solzhenitsyn’s novel
Cancer Ward, Pavel Nikolayevich Rusanov, a youthful Russian in his midforties, discovers that he has a tumor in his neck and is immediately whisked away into a cancer ward in some nameless hospital in the frigid north. The diagnosis of cancer—not the disease, but the mere stigma of its presence—becomes a death sentence for Rusanov. The illness strips him of his identity. It dresses him in a patient’s smock (a tragicomically cruel costume, no less blighting than a prisoner’s jumpsuit) and assumes absolute control of his actions. To be diagnosed with cancer, Rusanov discovers, is to enter a borderless medical gulag, a state even more invasive and paralyzing than the one that he has left behind. (Solzhenitsyn may have intended his absurdly totalitarian cancer hospital to parallel the absurdly totalitarian state outside it, yet when I once asked a woman with invasive cervical cancer about the parallel, she said sardonically, “Unfortunately, I did not need any metaphors to read the book. The cancer ward was my confining state, my prison.”)
As a doctor learning to tend cancer patients, I had only a partial glimpse of this confinement. But even skirting its periphery, I could still feel its power—the dense, insistent gravitational tug that pulls everything and everyone into the orbit of cancer. A colleague, freshly out of his fellowship, pulled me aside on my first week to offer some advice. “It’s called an immersive training program,” he said, lowering his voice. “But by immersive, they really mean drowning. Don’t let it work its way into everything you do. Have a life outside the hospital. You’ll need it, or you’ll get swallowed.”
But it was impossible not to be swallowed. In the parking lot of the hospital, a chilly, concrete box lit by neon floodlights, I spent the end of every evening after rounds in stunned incoherence, the car radio crackling vacantly in the background, as I compulsively tried to reconstruct the events of the day. The stories of my patients consumed me, and the decisions that I made haunted me. Was it worthwhile continuing yet another round of chemotherapy on a sixty-six-year-old pharmacist with lung cancer who had failed all other drugs? Was is better to try a tested and potent combination of drugs on a twenty-six-year-old woman with Hodgkin’s disease and risk losing her fertility, or to choose a more experimental combination that might spare it? Should a Spanish-speaking mother of three with colon cancer be enrolled in a new clinical trial when she can barely read the formal and inscrutable language of the consent forms?
Immersed in the day-to-day management of cancer, I could only see the lives and fates of my patients played out in color-saturated detail, like a television with the contrast turned too high. I could not pan back from the screen. I knew instinctively that these experiences were part of a much larger battle against cancer, but its contours lay far outside my reach. I had a novice’s hunger for history, but also a novice’s inability to envision it.


But as I emerged from the strange desolation of those two fellowship years, the questions about the larger story of cancer emerged with urgency: How old is cancer? What are the roots of our battle against this disease? Or, as patients often asked me: Where are we in the “war” on cancer? How did we get here? Is there an end? Can this war even be won?
This book grew out of the attempt to answer these questions. I delved into the history of cancer to give shape to the shape-shifting illness that I was confronting. I used the past to explain the present. The isolation and rage of a thirty-six-year-old woman with stage III breast cancer had ancient echoes in Atossa, the Persian queen
who swaddled her diseased breast in cloth to hide it and then, in a fit of nihilistic and prescient fury, had a slave cut it off with a knife. A patient’s desire to amputate her stomach, ridden with cancer—“sparing nothing,” as she put it to me—carried the memory of the perfection-obsessed nineteenth-century surgeon William Halsted, who had chiseled away at cancer with larger and more disfiguring surgeries, all in the hopes that cutting more would mean curing more.
Roiling underneath these medical, cultural, and metaphorical interceptions of cancer over the centuries was the biological understanding of the illness—an understanding that had morphed, often radically, from decade to decade. Cancer, we now know, is a disease caused by the uncontrolled growth of a single cell. This growth is unleashed by mutations—changes in DNA that specifically affect genes that incite unlimited cell growth. In a normal cell, powerful genetic circuits regulate cell division and cell death. In a cancer cell, these circuits have been broken, unleashing a cell that cannot stop growing.
That this seemingly simple mechanism—cell growth without barriers—can lie at the heart of this grotesque and multifaceted illness is a testament to the unfathomable power of cell growth. Cell division allows us as organisms to grow, to adapt, to recover, to repair—to live. And distorted and unleashed, it allows cancer cells to grow, to flourish, to adapt, to recover, and to repair—to live at the cost of our living. Cancer cells grow faster, adapt better. They are more perfect versions of ourselves.
The secret to battling cancer, then, is to find means to prevent these mutations from occurring in susceptible cells, or to find means to eliminate the mutated cells without compromising normal growth. The conciseness of that statement belies the enormity of the task. Malignant growth and normal growth are so genetically intertwined that unbraiding the two might be one of the most significant scientific challenges faced by our species. Cancer is built into our genomes: the genes that unmoor normal cell division are not foreign to our bodies, but rather mutated, distorted versions of the very genes that perform vital cellular functions. And cancer is imprinted in our society: as we extend our life span as a species, we inevitably unleash malignant growth (mutations in cancer genes accumulate with aging; cancer is thus intrinsically related to age). If we seek immortality, then so, too, in a rather perverse sense, does the cancer cell.
How, precisely, a future generation might learn to separate the entwined strands of normal growth from malignant growth remains a mystery. (“The universe,” the twentieth-century biologist
J. B. S. Haldane liked to say, “is not only queerer than we suppose, but queerer than we can suppose”—and so is the trajectory of science.) But this much is certain: the story, however it plays out, will contain indelible kernels of the past. It will be a story of inventiveness, resilience, and perseverance against what one writer called the most “relentless and insidious enemy” among human diseases. But it will also be a story of hubris, arrogance, paternalism, misperception, false hope, and hype, all leveraged against an illness that was just three decades ago widely touted as being “curable” within a few years.


In the bare hospital room ventilated by sterilized air, Carla was fighting her own war on cancer. When I arrived, she was sitting with peculiar calm on her bed, a schoolteacher jotting notes. (“But what notes?” she would later recall. “I just wrote and rewrote the same thoughts.”) Her mother, red-eyed and tearful, just off an overnight flight, burst into the room and then sat silently in a chair by the window, rocking forcefully. The din of activity around Carla had become almost a blur: nurses shuttling fluids in and out, interns donning masks and gowns, antibiotics being hung on IV poles to be dripped into her veins.
I explained the situation as best I could. Her day ahead would be full of tests, a hurtle from one lab to another. I would draw a bone marrow sample. More tests would be run by pathologists. But the preliminary tests suggested that Carla had acute lymphoblastic leukemia. It is one of the most common forms of cancer in children, but rare in adults. And it is—I paused here for emphasis, lifting my eyes up—often curable.
Curable. Carla nodded at that word, her eyes sharpening. Inevitable questions hung in the room: How curable? What were the chances that she would survive? How long would the treatment take? I laid out the odds. Once the diagnosis had been confirmed, chemotherapy would begin immediately and last more than one year. Her chances of being cured were about 30 percent, a little less than one in three.
We spoke for an hour, perhaps longer. It was now nine thirty in the morning. The city below us had stirred fully awake. The door shut behind me as I left, and a whoosh of air blew me outward and sealed Carla in.
Part One “Of blacke cholor, without boyling” (#ulink_4ecd16ce-5f0a-5072-abf7-0f61e24a7638)
In solving a problem of this sort
, the grand thing is to be able to reason backwards. That is a very useful accomplishment, and a very easy one, but people do not practice it much.
—Sherlock Holmes, in Sir Arthur Conan Doyle’s A Study in Scarlet
“A suppuration of blood” (#ulink_0cc9dce3-b2b1-516c-8c15-fd5c6fd10af2)

Physicians of the Utmost Fame

Were called at once; but when they came
They answered, as they took their Fees,
“There is no Cure for this Disease.”
—Hilaire Belloc
Its palliation is a daily task
, its cure a fervent hope.
—William Castle, describing leukemia in 1950
In a damp
fourteen-by-twenty-foot laboratory in Boston on a December morning in 1947, a man named Sidney Farber waited impatiently for the arrival of a parcel from New York. The “laboratory” was little more than a chemist’s closet, a poorly ventilated room buried in a half-basement of the Children’s Hospital, almost thrust into its back alley. A few hundred feet away, the hospital’s medical wards were slowly thrumming to work. Children in white smocks moved restlessly on small wrought-iron cots. Doctors and nurses shuttled busily between the rooms, checking charts, writing orders, and dispensing medicines. But Farber’s lab was listless and empty, a bare warren of chemicals and glass jars connected to the main hospital through a series of icy corridors. The sharp stench of embalming formalin wafted through the air. There were no patients in the rooms here, just the bodies and tissues of patients brought down through the tunnels for autopsies and examinations. Farber was a pathologist. His job involved dissecting specimens, performing autopsies, identifying cells, and diagnosing diseases, but never treating patients.
Farber’s specialty was pediatric pathology
, the study of children’s diseases. He had spent nearly twenty years in these subterranean rooms staring obsessively down his microscope and climbing through the academic ranks to become chief of pathology at Children’s. But for Farber, pathology was becoming a disjunctive form of medicine, a discipline more preoccupied with the dead than with the living. Farber now felt impatient watching illness from its sidelines, never touching or treating a live patient. He was tired of tissues and cells. He felt trapped, embalmed in his own glassy cabinet.
And so, Farber had decided to make a drastic professional switch. Instead of squinting at inert specimens under his lens, he would try to leap into the life of the clinics upstairs—from the microscopic world that he knew so well into the magnified real world of patients and illnesses. He would try to use the knowledge he had gathered from his pathological specimens to devise new therapeutic interventions. The parcel from New York contained a few vials of a yellow crystalline chemical named aminopterin. It had been shipped to his laboratory in Boston on the slim hope that it might halt the growth of leukemia in children.


Had Farber asked any of the pediatricians circulating in the wards above him about the likelihood of developing an antileukemic drug, they would have advised him not to bother trying. Childhood leukemia had fascinated, confused, and frustrated doctors for more than a century. The disease had been analyzed, classified, subclassified, and subdivided meticulously; in the musty, leatherbound books on the library shelves at Children’s—Anderson’s Pathology or Boyd’s Pathology of Internal Diseases—page upon page was plastered with images of leukemia cells and appended with elaborate taxonomies to describe the cells. Yet all this knowledge only amplified the sense of medical helplessness. The disease had turned into an object of empty fascination—a wax-museum doll—studied and photographed in exquisite detail but without any therapeutic or practical advances. “It gave physicians plenty to wrangle over
at medical meetings,” an oncologist recalled, “but it did not help their patients at all.” A patient with acute leukemia was brought to the hospital in a flurry of excitement, discussed on medical rounds with professorial grandiosity, and then, as a medical magazine drily noted, “diagnosed, transfused—and sent home to die.”

The study of leukemia had been mired in confusion and despair ever since its discovery. On March 19, 1845, a Scottish physician, John Bennett, had described an unusual case, a twenty-eight-year-old slate-layer with a mysterious swelling in his spleen. “He is of dark complexion,”
Bennett wrote of his patient, “usually healthy and temperate; [he] states that twenty months ago, he was affected with great listlessness on exertion, which has continued to this time. In June last he noticed a tumor in the left side of his abdomen which has gradually increased in size till four months since, when it became stationary.”
The slate-layer’s tumor might have reached its final, stationary point, but his constitutional troubles only accelerated. Over the next few weeks, Bennett’s patient spiraled from symptom to symptom—fevers, flashes of bleeding, sudden fits of abdominal pain—gradually at first, then on a tighter, faster arc, careening from one bout to another. Soon the slate-layer was on the verge of death with more swollen tumors sprouting in his armpits, his groin, and his neck. He was treated with the customary leeches and purging, but to no avail. At the autopsy a few weeks later, Bennett was convinced that he had found the reason behind the symptoms. His patient’s blood was chock-full of white blood cells. (White blood cells, the principal constituent of pus, typically signal the response to an infection, and Bennett reasoned that the slate-layer had succumbed to one.) “The following case seems to me particularly valuable,” he wrote self-assuredly, “as it will serve to demonstrate the existence of true pus, formed universally within the vascular system.”*
It would have been a perfectly satisfactory explanation except that Bennett could not find a source for the pus. During the necropsy, he pored carefully through the body, combing the tissues and organs for signs of an abscess or wound. But no other stigmata of infection were to be found. The blood had apparently spoiled—suppurated—of its own will, combusted spontaneously into true pus. “A suppuration of blood,” Bennett called his case. And he left it at that.
Bennett was wrong, of course, about his spontaneous “suppuration” of blood. A little over four months after Bennett had described the slater’s illness, a twenty-four-year-old German researcher, Rudolf Virchow, independently published
a case report with striking similarities to Bennett’s case. Virchow’s patient was a cook in her midfifties. White cells had explosively overgrown her blood, forming dense and pulpy pools in her spleen. At her autopsy, pathologists had likely not even needed a microscope to distinguish the thick, milky layer of white cells floating above the red.
Virchow, who knew of Bennett’s case, couldn’t bring himself to believe Bennett’s theory. Blood, Virchow argued, had no reason to transform impetuously into anything. Moreover, the unusual symptoms bothered him: What of the massively enlarged spleen? Or the absence of any wound or source of pus in the body? Virchow began to wonder if the blood itself was abnormal. Unable to find a unifying explanation for it, and seeking a name for this condition
, Virchow ultimately settled for weisses Blut—white blood—no more than a literal description of the millions of white cells he had seen under his microscope. In 1847, he changed the name to the more academic-sounding “leukemia”—from leukos, the Greek word for “white.”


Renaming the disease—from the florid “suppuration of blood” to the flat weisses Blut—hardly seems like an act of scientific genius, but it had a profound impact on the understanding of leukemia. An illness, at the moment of its discovery, is a fragile idea, a hothouse flower—deeply, disproportionately influenced by names and classifications. (More than a century later, in the early 1980s, another change in name
—from gay related immune disease (GRID) to acquired immuno deficiency syndrome (AIDS)—would signal an epic shift in the understanding of that disease.*) Like Bennett, Virchow didn’t understand leukemia. But unlike Bennett, he didn’t pretend to understand it. His insight lay entirely in the negative. By wiping the slate clean of all preconceptions, he cleared the field for thought.
The humility of the name (and the underlying humility about his understanding of cause) epitomized Virchow’s approach to medicine
. As a young professor at the University of Würzburg, Virchow’s work soon extended far beyond naming leukemia. A pathologist by training, he launched a project that would occupy him for his life: describing human diseases in simple cellular terms.
It was a project born of frustration. Virchow entered medicine in the early 1840s, when nearly every disease was attributed to the workings of some invisible force: miasmas, neuroses, bad humors, and hysterias. Perplexed by what he couldn’t see, Virchow turned with revolutionary zeal to what he could see: cells under the microscope. In 1838, Matthias Schleiden, a botanist, and Theodor Schwann, a physiologist, both working in Germany, had claimed that all living organisms were built out of fundamental building blocks called cells. Borrowing and extending this idea, Virchow set out to create a “cellular theory” of human biology, basing it on two fundamental tenets. First, that human bodies (like the bodies of all animals and plants) were made up of cells. Second, that cells only arose from other cells—omnis cellula e cellula, as he put it.
The two tenets might have seemed simplistic, but they allowed Virchow to propose a crucially important hypothesis about the nature of human growth. If cells only arose from other cells, then growth could occur in only two ways: either by increasing cell numbers or by increasing cell size. Virchow called these two modes hyperplasia and hypertrophy. In hypertrophy, the number of cells did not change; instead, each individual cell merely grew in size—like a balloon being blown up. Hyperplasia, in contrast, was growth by virtue of cells increasing in number. Every growing human tissue could be described in terms of hypertrophy and hyperplasia. In adult animals, fat and muscle usually grow by hypertrophy. In contrast, the liver, blood, the gut, and the skin all grow through hyperplasia—cells becoming cells becoming more cells, omnis cellula e cellula e cellula.
That explanation was persuasive, and it provoked a new understanding not just of normal growth, but of pathological growth as well. Like normal growth, pathological growth could also be achieved through hypertrophy and hyperplasia. When the heart muscle is forced to push against a blocked aortic outlet, it often adapts by making every muscle cell bigger to generate more force, eventually resulting in a heart so overgrown that it may be unable to function normally—pathological hypertrophy.
Conversely, and importantly for this story, Virchow soon stumbled upon the quintessential disease of pathological hyperplasia—cancer. Looking at cancerous growths through his microscope, Virchow discovered an uncontrolled growth of cells—hyperplasia in its extreme form. As Virchow examined the architecture of cancers, the growth often seemed to have acquired a life of its own, as if the cells had become possessed by a new and mysterious drive to grow. This was not just ordinary growth, but growth redefined, growth in a new form. Presciently (although oblivious of the mechanism) Virchow called it neoplasia—novel, inexplicable, distorted growth, a word that would ring through the history of cancer.*
By the time Virchow died in 1902, a new theory of cancer had slowly coalesced out of all these observations. Cancer was a disease of pathological hyperplasia in which cells acquired an autonomous will to divide. This aberrant, uncontrolled cell division created masses of tissue (tumors) that invaded organs and destroyed normal tissues. These tumors could also spread from one site to another, causing outcroppings of the disease—called metastases—in distant sites, such as the bones, the brain, or the lungs. Cancer came in diverse forms—breast, stomach, skin, and cervical cancer, leukemias and lymphomas. But all these diseases were deeply connected at the cellular level. In every case, cells had all acquired the same characteristic: uncontrollable pathological cell division.
With this understanding, pathologists who studied leukemia in the late 1880s now circled back to Virchow’s work. Leukemia, then, was not a suppuration of blood, but neoplasia of blood. Bennett’s earlier fantasy
had germinated an entire field of fantasies among scientists, who had gone searching (and dutifully found) all sorts of invisible parasites and bacteria bursting out of leukemia cells. But once pathologists stopped looking for infectious causes and refocused their lenses on the disease, they discovered the obvious analogies between leukemia cells and cells of other forms of cancer. Leukemia was a malignant proliferation of white cells in the blood. It was cancer in a molten, liquid form.
With that seminal observation, the study of leukemias suddenly found clarity and spurted forward. By the early 1900s, it was clear that the disease came in several forms. It could be chronic and indolent, slowly choking the bone marrow and spleen, as in Virchow’s original case (later termed chronic leukemia). Or it could be acute and violent, almost a different illness in its personality, with flashes of fever, paroxysmal fits of bleeding, and a dazzlingly rapid overgrowth of cells—as in Bennett’s patient.
This second version of the disease, called acute leukemia, came in two further subtypes, based on the type of cancer cell involved. Normal white cells in the blood can be broadly divided into two types of cells—myeloid cells or lymphoid cells. Acute myeloid leukemia (AML) was a cancer of the myeloid cells. Acute lymphoblastic leukemia (ALL) was cancer of immature lymphoid cells. (Cancers of more mature lymphoid cells are called lymphomas.)
In children, leukemia was most commonly ALL—lymphoblastic leukemia—and was almost always swiftly lethal. In 1860, a student of Virchow’s, Michael Anton Biermer, described
the first known case of this form of childhood leukemia. Maria Speyer, an energetic, vivacious, and playful five-year-old daughter of a Würzburg carpenter, was initially seen at the clinic because she had become lethargic in school and developed bloody bruises on her skin. The next morning, she developed a stiff neck and a fever, precipitating a call to Biermer for a home visit. That night, Biermer drew a drop of blood from Maria’s veins, looked at the smear using a candlelit bedside microscope, and found millions of leukemia cells in the blood. Maria slept fitfully late into the evening. Late the next afternoon, as Biermer was excitedly showing his colleagues the specimens of “exquisit Fall von Leukämie” (an exquisite case of leukemia), Maria vomited bright red blood and lapsed into a coma. By the time Biermer returned to her house that evening, the child had been dead for several hours. From its first symptom to diagnosis to death
, her galloping, relentless illness had lasted no more than three days.


Although nowhere as aggressive as Maria Speyer’s leukemia, Carla’s illness was astonishing in its own right. Adults, on average, have about five thousand white blood cells circulating per milliliter of blood. Carla’s blood contained ninety thousand cells per milliliter—nearly twentyfold the normal level. Ninety-five percent of these cells were blasts—malignant lymphoid cells produced at a frenetic pace but unable to mature into fully developed lymphocytes. In acute lymphoblastic leukemia, as in some other cancers, the overproduction of cancer cells is combined with a mysterious arrest in the normal maturation of cells. Lymphoid cells are thus produced in vast excess, but, unable to mature, they cannot fulfill their normal function in fighting microbes. Carla had immunological poverty in the face of plenty.
White blood cells are produced in the bone marrow. Carla’s bone marrow biopsy, which I saw under the microscope the morning after I first met her, was deeply abnormal. Although superficially amorphous, bone marrow is a highly organized tissue—an organ, in truth—that generates blood in adults. Typically, bone marrow biopsies contain spicules of bone and, within these spicules, islands of growing blood cells—nurseries for the genesis of new blood. In Carla’s marrow, this organization had been fully destroyed. Sheet upon sheet of malignant blasts packed the marrow space, obliterating all anatomy and architecture, leaving no space for any production of blood.
Carla was at the edge of a physiological abyss. Her red cell count had dipped so low that her blood was unable to carry its full supply of oxygen (her headaches, in retrospect, were the first sign of oxygen deprivation). Her platelets, the cells responsible for clotting blood, had collapsed to nearly zero, causing her bruises.
Her treatment would require extraordinary finesse. She would need chemotherapy to kill her leukemia, but the chemotherapy would collaterally decimate any remnant normal blood cells. We would push her deeper into the abyss to try to rescue her. For Carla, the only way out would be the way through.


Sidney Farber was born in Buffalo, New York, in 1903, one year after Virchow’s death in Berlin. His father, Simon Farber, a former bargeman in Poland, had immigrated to America in the late nineteenth century and worked in an insurance agency. The family lived in modest circumstances at the eastern edge of town, in a tight-knit, insular, and often economically precarious Jewish community of shop owners, factory workers, bookkeepers, and peddlers. Pushed relentlessly to succeed, the Farber children were held to high academic standards. Yiddish was spoken upstairs, but only German and English were allowed downstairs. The elder Farber often brought home textbooks and scattered them across the dinner table, expecting each child to select and master one book, then provide a detailed report for him.
Sidney, the third of fourteen children, thrived in this environment of high aspirations. He studied both biology and philosophy in college and graduated from the University of Buffalo in 1923, playing the violin at music halls to support his college education. Fluent in German, he trained in medicine at Heidelberg and Freiburg, then, having excelled in Germany, found a spot as a second-year medical student at Harvard Medical School in Boston. (The circular journey from New York to Boston via Heidelberg was not unusual. In the mid-1920s, Jewish students often found it impossible to secure medical-school spots in America—often succeeding in European, even German, medical schools before returning to study medicine in their native country.) Farber thus arrived at Harvard as an outsider. His colleagues found him arrogant and insufferable, but, he too, relearning lessons that he had already learned, seemed to be suffering through it all. He was formal, precise, and meticulous, starched in his appearance and his mannerisms and commanding in presence. He was promptly nicknamed Four-Button Sid for his propensity for wearing formal suits to his classes.
Farber completed his advanced training
in pathology in the late 1920s and became the first full-time pathologist at the Children’s Hospital in Boston. He wrote a marvelous study on the classification of children’s tumors and a textbook, The Postmortem Examination, widely considered a classic in the field. By the mid-1930s, he was firmly ensconced in the back alleys of the hospital as a preeminent pathologist—a “doctor of the dead.”
Yet the hunger to treat patients still drove Farber. And sitting in his basement laboratory in the summer of 1947, Farber had a single inspired idea: he chose, among all cancers, to focus his attention on one of its oddest and most hopeless variants—childhood leukemia. To understand cancer as a whole, he reasoned, you needed to start at the bottom of its complexity, in its basement. And despite its many idiosyncrasies, leukemia possessed a singularly attractive feature: it could be measured.
Science begins with counting. To understand a phenomenon, a scientist must first describe it; to describe it objectively, he must first measure it. If cancer medicine was to be transformed into a rigorous science, then cancer would need to be counted somehow—measured in some reliable, reproducible way.
In this, leukemia was different from nearly every other type of cancer. In a world before CT scans and MRIs, quantifying the change in size of an internal solid tumor in the lung or the breast was virtually impossible without surgery: you could not measure what you could not see. But leukemia, floating freely in the blood, could be measured as easily as blood cells—by drawing a sample of blood or bone marrow and looking at it under a microscope.
If leukemia could be counted, Farber reasoned, then any intervention—a chemical sent circulating through the blood, say—could be evaluated for its potency in living patients. He could watch cells grow or die in the blood and use that to measure the success or failure of a drug. He could perform an “experiment” on cancer.
The idea mesmerized Farber. In the 1940s and ’50s, young biologists were galvanized by the idea of using simple models to understand complex phenomena. Complexity was best understood by building from the ground up. Single-celled organisms such as bacteria would reveal the workings of massive, multicellular animals such as humans. What is true for E. coli
[a microscopic bacterium], the French biochemist Jacques Monod would grandly declare in 1954, must also be true for elephants.
For Farber, leukemia epitomized this biological paradigm. From this simple, atypical beast he would extrapolate into the vastly more complex world of other cancers; the bacterium would teach him to think about the elephant. He was, by nature, a quick and often impulsive thinker. And here, too, he made a quick, instinctual leap. The package from New York was waiting in his laboratory that December morning. As he tore it open, pulling out the glass vials of chemicals, he scarcely realized that he was throwing open an entirely new way of thinking about cancer.
“A monster more insatiable than the guillotine” (#ulink_ee8c0dac-6081-5abb-bdc0-b756f3746163)
The medical importance of leukemia
has always been disproportionate to its actual incidence. . . . Indeed, the problems encountered in the systemic treatment of leukemia were indicative of the general directions in which cancer research as a whole was headed.
—Jonathan Tucker, Ellie: A Child’s Fight Against Leukemia
There were few successes in the treatment
of disseminated cancer. . . . It was usually a matter of watching the tumor get bigger, and the patient, progressively smaller.
—John Laszlo, The Cure of Childhood Leukemia: Into the Age of Miracles
Sidney Farber’s package of chemicals happened to arrive at a particularly pivotal moment in the history of medicine. In the late 1940s, a cornucopia of pharmaceutical discoveries
was tumbling open in labs and clinics around the nation. The most iconic of these new drugs were the antibiotics. Penicillin, that precious chemical that had to be milked to its last droplet during World War II (in 1939, the drug was reextracted
from the urine of patients who had been treated with it to conserve every last molecule), was by the early fifties being produced in thousand-gallon vats. In 1942, when Merck had shipped
out its first batch of penicillin—a mere five and a half grams of the drug—that amount had represented half of the entire stock of the antibiotic in America. A decade later, penicillin
was being mass-produced so effectively that its price had sunk to four cents for a dose, one-eighth the cost of a half gallon of milk.
New antibiotics followed in the footsteps of penicillin: chloramphenicol in 1947
, tetracycline in 1948
. In the winter of 1949, when yet another miraculous antibiotic, streptomycin, was purified out of a clod of mold from a chicken farmer’s barnyard, Time magazine splashed the phrase “The remedies are in our own backyard,”
prominently across its cover. In a brick building on the far corner
of Children’s Hospital, in Farber’s own backyard, a microbiologist named John Enders was culturing poliovirus in rolling plastic flasks, the first step that culminated in the development of the Sabin and Salk polio vaccines. New drugs appeared at an astonishing rate: by 1950, more than half the medicines
in common medical use had been unknown merely a decade earlier.
Perhaps even more significant than these miracle drugs, shifts in public health and hygiene also drastically altered the national physiognomy of illness. Typhoid fever
, a contagion whose deadly swirl could decimate entire districts in weeks, melted away as the putrid water supplies of several cities were cleansed by massive municipal efforts. Even tuberculosis
, the infamous “white plague” of the nineteenth century, was vanishing, its incidence plummeting by more than half between 1910 and 1940, largely due to better sanitation and public hygiene efforts. The life expectancy of Americans
rose from forty-seven to sixty-eight in half a century, a greater leap in longevity than had been achieved over several previous centuries.
The sweeping victories of postwar medicine illustrated the potent and transformative capacity of science and technology in American life. Hospitals proliferated
—between 1945 and 1960, nearly one thousand new hospitals were launched nationwide; between 1935 and 1952, the number of patients admitted more than doubled from 7 million to 17 million per year. And with the rise in medical care came the concomitant expectation of medical cure. As one student observed,
“When a doctor has to tell a patient that there is no specific remedy for his condition, [the patient] is apt to feel affronted, or to wonder whether the doctor is keeping abreast of the times.”
In new and sanitized suburban towns, a young generation thus dreamed of cures—of a death-free, disease-free existence. Lulled by the idea of the durability
of life, they threw themselves into consuming durables: boat-size Studebakers, rayon leisure suits, televisions, radios, vacation homes, golf clubs, barbecue grills, washing machines. In Levittown, a sprawling suburban settlement built in a potato field on Long Island—a symbolic utopia—“illness” now ranked third
in a list of “worries,” falling behind “finances” and “child-rearing.” In fact, rearing children was becoming a national preoccupation at an unprecedented level. Fertility rose steadily
—by 1957, a baby was being born every seven seconds in America. The “affluent society,”
as the economist John Galbraith described it, also imagined itself as eternally young, with an accompanying guarantee of eternal health—the invincible society.


But of all diseases, cancer had refused to fall into step in this march of progress. If a tumor was strictly local (i.e., confined to a single organ or site so that it could be removed by a surgeon), the cancer stood a chance of being cured. Extirpations, as these procedures came to be called, were a legacy of the dramatic advances of nineteenth-century surgery. A solitary malignant lump in the breast, say, could be removed via a radical mastectomy pioneered by the great surgeon William Halsted at Johns Hopkins in the 1890s. With the discovery of X-rays in the early 1900s, radiation could also be used to kill tumor cells at local sites.
But scientifically, cancer still remained a black box, a mysterious entity that was best cut away en bloc rather than treated by some deeper medical insight. To cure cancer (if it could be cured at all), doctors had only two strategies: excising the tumor surgically or incinerating it with radiation—a choice between the hot ray and the cold knife.
In May 1937
, almost exactly a decade before Farber began his experiments with chemicals, Fortune magazine published what it called a “panoramic survey” of cancer medicine. The report was far from comforting: “The startling fact is that no new principle of treatment, whether for cure or prevention, has been introduced. . . . The methods of treatment have become more efficient and more humane. Crude surgery without anesthesia or asepsis has been replaced by modern painless surgery with its exquisite technical refinement. Biting caustics that ate into the flesh of past generations of cancer patients have been obsolesced by radiation with X-ray and radium. . . . But the fact remains that the cancer ‘cure’ still includes only two principles—the removal and destruction of diseased tissue [the former by surgery; the latter by X-rays]. No other means have been proved.”
The Fortune article was titled “Cancer: The Great Darkness,” and the “darkness,” the authors suggested, was as much political as medical. Cancer medicine was stuck in a rut not only because of the depth of medical mysteries that surrounded it, but because of the systematic neglect of cancer research: “There are not over two dozen funds in the U.S. devoted to fundamental cancer research. They range in capital from about $500 up to about $2,000,000, but their aggregate capitalization is certainly not much more than $5,000,000. . . . The public willingly spends a third of that sum in an afternoon to watch a major football game.”
This stagnation of research funds stood in stark contrast to the swift rise to prominence of the disease itself. Cancer had certainly been present and noticeable in nineteenth-century America, but it had largely lurked in the shadow of vastly more common illnesses. In 1899, when Roswell Park
, a well-known Buffalo surgeon, had argued that cancer would someday overtake smallpox, typhoid fever, and tuberculosis to become the leading cause of death in the nation, his remarks had been perceived as a rather “startling prophecy,” the hyperbolic speculations of a man who, after all, spent his days and nights operating on cancer. But by the end of the decade, Park’s remarks were becoming less and less startling, and more and more prophetic by the day. Typhoid, aside from a few scattered outbreaks, was becoming increasingly rare. Smallpox was on the decline
; by 1949, it would disappear from America altogether. Meanwhile cancer was already outgrowing other diseases, ratcheting its way up the ladder of killers. Between 1900 and 1916
, cancer-related mortality grew by 29.8 percent, edging out tuberculosis as a cause of death. By 1926, cancer
had become the nation’s second most common killer, just behind heart disease.
“Cancer: The Great Darkness” wasn’t alone in building a case for a coordinated national response to cancer. In May that year,
Life carried its own dispatch on cancer research, which conveyed the same sense of urgency. The New York Times published two reports on rising cancer rates, in April and June. When cancer appeared
in the pages of Time in July 1937, interest in what was called the “cancer problem” was like a fierce contagion in the media.


Proposals to mount a systematic national response against cancer had risen and ebbed rhythmically in America since the early 1900s. In 1907, a group of cancer surgeons had congregated at the New Willard Hotel in Washington to create an organization to lobby Congress for more funds for cancer research. By 1910, this organization, the American Association for Cancer Research
, had convinced President Taft to propose to Congress a national laboratory dedicated to cancer research. But despite initial interest in the plan, the efforts had stalled in Washington after a few fitful attempts, largely because of a lack of political support.
In the late 1920s, a decade after Taft’s proposal had been tabled, cancer research found a new and unexpected champion—Matthew Neely, a dogged and ebullient former lawyer from Fairmont, West Virginia, serving his first term in the Senate. Although Neely had relatively little experience in the politics of science, he had noted the marked increase in cancer mortality in the previous decade—from 70,000 men and women in 1911
to 115,000 in 1927. Neely asked Congress
to advertise a reward of $5 million for any “information leading to the arrest of human cancer.”
It was a lowbrow strategy—the scientific equivalent of hanging a mug shot in a sheriff’s office—and it generated a reflexively lowbrow response. Within a few weeks
, Neely’s office in Washington was flooded with thousands of letters from quacks and faith healers purporting every conceivable remedy for cancer: rubs, tonics, ointments, anointed handkerchiefs, salves, and blessed water. Congress, exasperated with the response, finally authorized $50,000 for Neely’s Cancer Control Bill, almost comically cutting its budget back to just 1 percent of the requested amount.
In 1937, the indefatigable Neely, reelected to the Senate, launched yet another effort to launch a national attack on cancer, this time jointly with Senator Homer Bone and Representative Warren Magnuson. By now, cancer had considerably magnified in the public eye. The Fortune and Time articles had fanned anxiety and discontent, and politicians were eager to demonstrate a concrete response. In June, a joint Senate-House conference
was held to craft legislation to address the issue. After initial hearings, the bill raced through Congress and was passed unanimously by a joint session on July 23, 1937. Two weeks later, on August 5, President Roosevelt signed the National Cancer Institute Act.
The act created a new scientific unit called the National Cancer Institute (NCI), designed to coordinate cancer research and education.* An advisory council of scientists
for the institute was assembled from universities and hospitals. A state-of-the-art laboratory space, with gleaming halls and conference rooms, was built among leafy arcades and gardens in suburban Bethesda, a few miles from the nation’s capital. “The nation is marshaling its forces
to conquer cancer, the greatest scourge that has ever assailed the human race,” Senator Bone announced reassuringly while breaking ground for the building on October 3, 1938. After nearly two decades of largely fruitless efforts, a coordinated national response to cancer seemed to be on its way at last.
All of this was a bold, brave step in the right direction—except for its timing. By the early winter of 1938, just months after the inauguration of the NCI campus in Bethesda, the battle against cancer was overshadowed by the tremors of a different kind of war. In November, Nazi troops embarked on a nationwide pogrom against Jews in Germany, forcing thousands into concentration camps. By late winter, military conflicts had broken out all over Asia and Europe, setting the stage for World War II. By 1939, those skirmishes had fully ignited, and in December 1941, America was drawn inextricably into the global conflagration.
The war necessitated a dramatic reordering of priorities. The U.S. Marine Hospital
in Baltimore, which the NCI had once hoped to convert into a clinical cancer center, was now swiftly reconfigured into a war hospital. Scientific research funding stagnated and was shunted into projects directly relevant to the war. Scientists, lobbyists, physicians, and surgeons fell off the public radar screen—“mostly silent,”
as one researcher recalled, “their contributions usually summarized in obituaries.”
An obituary might as well have been written for the National Cancer Institute. Congress’s promised funds for a “programmatic response to cancer”
never materialized, and the NCI languished in neglect. Outfitted with every modern facility imaginable in the 1940s, the institute’s sparkling campus turned into a scientific ghost town. One scientist jokingly called it “a nice quiet place out here in the country.
In those days,” the author continued, “it was pleasant to drowse under the large, sunny windows.”*
The social outcry about cancer also drifted into silence. After the brief flurry of attention in the press, cancer again became the great unmentionable, the whispered-about disease that no one spoke about publicly. In the early 1950s, Fanny Rosenow
, a breast cancer survivor and cancer advocate, called the New York Times to post an advertisement for a support group for women with breast cancer. Rosenow was put through, puzzlingly, to the society editor of the newspaper. When she asked about placing her announcement, a long pause followed. “I’m sorry, Ms. Rosenow, but the Times cannot publish the word breast or the word cancer in its pages.

“Perhaps,” the editor continued, “you could say there will be a meeting about diseases of the chest wall.”
Rosenow hung up, disgusted.


When Farber entered the world of cancer in 1947, the public outcry of the past decade had dissipated. Cancer had again become a politically silent illness. In the airy wards of the Children’s Hospital, doctors and patients fought their private battles against cancer. In the tunnels downstairs, Farber fought an even more private battle with his chemicals and experiments.
This isolation was key to Farber’s early success. Insulated from the spotlights of public scrutiny, he worked on a small, obscure piece of the puzzle. Leukemia was an orphan disease, abandoned by internists, who had no drugs to offer for it, and by surgeons, who could not possibly operate on blood. “Leukemia,” as one physician put it
, “in some senses, had not [even] been cancer before World War II.” The illness lived on the borderlands of illnesses, a pariah lurking between disciplines and departments—not unlike Farber himself.
If leukemia “belonged” anywhere
, it was within hematology, the study of normal blood. If a cure for it was to be found, Farber reasoned, it would be found by studying blood. If he could uncover how normal blood cells were generated, he might stumble backward into a way to block the growth of abnormal leukemic cells. His strategy, then, was to approach the disease from the normal to the abnormal—to confront cancer in reverse.
Much of what Farber knew about normal blood he had learned from George Minot. A thin, balding aristocrat with pale, intense eyes, Minot ran a laboratory in a colonnaded, brick-and-stone structure off Harrison Avenue in Boston, just a few miles down the road from the sprawling hospital complex on Longwood Avenue that included Children’s Hospital. Like many hematologists at Harvard, Farber had trained briefly with Minot in the 1920s before joining the staff at Children’s.
Every decade has a unique hematological riddle, and for Minot’s era, that riddle was pernicious anemia. Anemia is the deficiency of red blood cells—and its most common form arises from a lack of iron, a crucial nutrient used to build red blood cells. But pernicious anemia, the rare variant that Minot studied, was not caused by iron deficiency (indeed, its name derives from its intransigence to the standard treatment of anemia with iron). By feeding patients increasingly macabre concoctions—half a pound of chicken liver
, half-cooked hamburgers, raw hog stomach, and even once the regurgitated gastric juices
of one of his students (spiced up with butter, lemon, and parsley
)—Minot and his team of researchers
conclusively demonstrated in 1926
that pernicious anemia was caused by the lack of a critical micronutrient, a single molecule later identified as vitamin B12. In 1934, Minot and two of his colleagues
won the Nobel Prize for this pathbreaking work. Minot had shown that replacing a single molecule could restore the normalcy of blood in this complex hematological disease. Blood was an organ whose activity could be turned on and off by molecular switches.
There was another form of nutritional anemia that Minot’s group had not tackled, an anemia just as “pernicious”—although in the moral sense of that word. Eight thousand miles away, in the cloth mills of Bombay
(owned by English traders and managed by their cutthroat local middlemen), wages had been driven to such low levels that the mill workers lived in abject poverty, malnourished and without medical care. When English physicians tested these mill workers in the 1920s to study the effects of this chronic malnutrition, they discovered that many of them, particularly women after childbirth, were severely anemic. (This was yet another colonial fascination: to create the conditions of misery in a population, then subject it to social or medical experimentation.)
In 1928, a young English physician named Lucy Wills,
freshly out of the London School of Medicine for Women, traveled on a grant to Bombay to study this anemia. Wills was an exotic among hematologists, an adventurous woman driven by a powerful curiosity about blood willing to travel to a faraway country to solve a mysterious anemia on a whim. She knew of Minot’s work. But unlike Minot’s anemia, she found that the anemia in Bombay couldn’t be reversed by Minot’s concoctions or by vitamin B12. Astonishingly, she found she could cure it with Marmite, the dark, yeasty spread then popular among health fanatics in England and Australia. Wills could not determine the key chemical nutrient of Marmite. She called it the Wills factor
.
Wills factor turned out to be folic acid, or folate, a vitamin-like substance found in fruits and vegetables (and amply in Marmite). When cells divide, they need to make copies of DNA—the chemical that carries all the genetic information in a cell. Folic acid is a crucial building block for DNA and is thus vital for cell division. Since blood cells are produced by arguably the most fearsome rate of cell division in the human body—more than 300 billion cells a day—the genesis of blood is particularly dependent on folic acid. In its absence (in men and women starved of vegetables, as in Bombay) the production of new blood cells in the bone marrow halts. Millions of half-matured cells spew out, piling up like half-finished goods bottlenecked in an assembly line. The bone marrow becomes a dysfunctional mill, a malnourished biological factory oddly reminiscent of the cloth factories of Bombay.


These links—between vitamins, bone marrow, and normal blood—kept Farber preoccupied in the early summer of 1946. In fact, his first clinical experiment, inspired by this very connection, turned into a horrific mistake. Lucy Wills had observed that folic acid, if administered to nutrient-deprived patients, could restore the normal genesis of blood. Farber wondered whether administering folic acid to children with leukemia might also restore normalcy to their blood. Following that tenuous trail, he obtained some synthetic folic acid, recruited a cohort of leukemic children, and started injecting folic acid into them.
In the months that passed, Farber found that folic acid, far from stopping the progression of leukemia, actually accelerated it. In one patient, the white cell count nearly doubled. In another, the leukemia cells exploded into the bloodstream and sent fingerlings of malignant cells to infiltrate the skin. Farber stopped the experiment in a hurry. He called this phenomenon acceleration
, evoking some dangerous object in free fall careering toward its end.
Pediatricians at Children’s Hospital were furious about Farber’s trial. The folate analogues had not just accelerated the leukemia; they had likely hastened the death of the children. But Farber was intrigued. If folic acid accelerated the leukemia cells in children, what if he could cut off its supply with some other drug—an antifolate? Could a chemical that blocked the growth of white blood cells stop leukemia?
The observations of Minot and Wills began to fit into a foggy picture. If the bone marrow was a busy cellular factory to begin with, then a marrow occupied with leukemia was that factory in overdrive, a deranged manufacturing unit for cancer cells. Minot and Wills had turned the production lines of the bone marrow on by adding nutrients to the body. But could the malignant marrow be shut off by choking the supply of nutrients? Could the anemia of the mill workers in Bombay be re-created therapeutically in the medical units of Boston?
In his long walks from his laboratory
under Children’s Hospital to his house on Amory Street in Brookline, Farber wondered relentlessly about such a drug. Dinner, in the dark-wood-paneled rooms of the house, was usually a sparse, perfunctory affair. His wife, Norma, a musician and writer, talked about the opera and poetry; Sidney, of autopsies, trials, and patients. As he walked back to the hospital at night, Norma’s piano tinkling practice scales in his wake, the prospect of an anticancer chemical haunted him. He imagined it palpably, visibly, with a fanatic’s enthusiasm. But he didn’t know what it was or what to call it. The word chemotherapy, in the sense we understand it today, had never been used for anticancer medicines.* The elaborate armamentarium of “antivitamins” that Farber had dreamed up so vividly in his fantasies did not exist.


Farber’s supply of folic acid for his disastrous first trial had come from the laboratory of an old friend, a chemist, Yellapragada Subbarao—or Yella, as most of his colleagues called him. Yella was a pioneer in many ways, a physician turned cellular physiologist, a chemist who had accidentally wandered into biology. His scientific meanderings had been presaged by more desperate and adventuresome physical meanderings. He had arrived in Boston in 1923
, penniless and unprepared, having finished his medical training in India and secured a scholarship for a diploma at the School of Tropical Health at Harvard. The weather in Boston, Yella discovered, was far from tropical. Unable to find a medical job in the frigid, stormy winter (he had no license to practice medicine in the United States), he started as a night porter at the Brigham and Women’s Hospital, opening doors, changing sheets, and cleaning urinals.
The proximity to medicine paid off. Subbarao made friends and connections at the hospital and switched to a day job as a researcher in the Division of Biochemistry. His initial project involved purifying molecules out of living cells, dissecting them chemically to determine their compositions—in essence, performing a biochemical “autopsy” on cells. The approach required more persistence than imagination, but it produced remarkable dividends. Subbarao purified a molecule called ATP, the source of energy in all living beings (ATP carries chemical “energy” in the cell), and another molecule called creatine, the energy carrier in muscle cells. Any one of these achievements should have been enough to guarantee him a professorship at Harvard. But Subbarao was a foreigner, a reclusive, nocturnal, heavily accented vegetarian who lived in a one-room apartment downtown, befriended only by other nocturnal recluses such as Farber. In 1940, denied tenure and recognition, Yella huffed off to join Lederle Labs, a pharmaceutical laboratory in upstate New York, owned by the American Cyanamid Corporation, where he had been asked to run a group on chemical synthesis.
At Lederle, Yella Subbarao quickly reformulated his old strategy and focused on making synthetic versions of the natural chemicals that he had found within cells, hoping to use them as nutritional supplements. In the 1920s, another drug company
, Eli Lilly, had made a fortune selling a concentrated form of vitamin B12, the missing nutrient in pernicious anemia. Subbarao decided to focus his attention on the other anemia, the neglected anemia of folate deficiency. But in 1946, after many failed attempts
to extract the chemical from pigs’ livers, he switched tactics and started to synthesize folic acid from scratch, with the help of a team of scientists including Harriet Kiltie, a young chemist at Lederle.
The chemical reactions to make folic acid brought a serendipitous bonus. Since the reactions had several intermediate steps, Subbarao and Kiltie could create variants of folic acid through slight alterations in the recipe. These variants of folic acid—closely related molecular mimics—possessed counterintuitive properties. Enzymes and receptors in cells typically work by recognizing molecules using their chemical structure. But a “decoy” molecular structure—one that nearly mimics the natural molecule—can bind to the receptor or enzyme and block its action, like a false key jamming a lock. Some of Yella’s molecular mimics could thus behave like antagonists to folic acid.
These were precisely the antivitamins that Farber had been fantasizing about. Farber wrote to Kiltie and Subbarao asking them if he could use their folate antagonists on patients with leukemia. Subbarao consented. In the late summer of 1947, the first package of antifolate left Lederle’s labs in New York and arrived in Farber’s laboratory.
Farber’s Gauntlet (#ulink_82ba2cda-1310-56f8-affb-2d4790da67fa)
Throughout the centuries
the sufferer from this disease has been the subject of almost every conceivable form of experimentation. The fields and forests, the apothecary shop and the temple, have been ransacked for some successful means of relief from this intractable malady. Hardly any animal has escaped making its contribution, in hair or hide, tooth or toenail, thymus or thyroid, liver or spleen, in the vain search by man for a means of relief.
—William Bainbridge
The search for a way to eradicate this scourge
. . . is left to incidental dabbling and uncoordinated research.
—The Washington Post, 1946
Seven miles south of the Longwood hospitals in Boston, the town of Dorchester is a typical sprawling New England suburb, a triangle wedged between the sooty industrial settlements to the west and the gray-green bays of the Atlantic to its east. In the late 1940s, waves of Jewish and Irish immigrants—shipbuilders, iron casters, railway engineers, fishermen, and factory workers—settled in Dorchester, occupying rows of brick-and-clapboard houses that snaked their way up Blue Hill Avenue. Dorchester reinvented itself as the quintessential suburban family town, with parks and playgrounds along the river, a golf course, a church, and a synagogue. On Sunday afternoons, families converged at Franklin Park to walk through its leafy pathways or to watch ostriches, polar bears, and tigers at its zoo.
On August 16, 1947, in a house across from the zoo, the child of a ship worker in the Boston yards fell mysteriously ill with a low-grade fever that waxed and waned over two weeks without pattern, followed by increasing lethargy and pallor. Robert Sandler
was two years old. His twin, Elliott, was an active, cherubic toddler in perfect health.
Ten days after his first fever, Robert’s condition worsened significantly. His temperature climbed higher. His complexion turned from rosy to a spectral milky white. He was brought to Children’s Hospital in Boston. His spleen, a fist-size organ that stores and makes blood (usually barely palpable underneath the rib cage), was visibly enlarged, heaving down like an overfilled bag. A drop of blood under Farber’s
microscope revealed the identity of his illness; thousands of immature lymphoid leukemic blasts were dividing in a frenzy, their chromosomes condensing and uncondensing, like tiny clenched and unclenched fists.
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 gamut 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 (#ulink_c86c90ad-ee1e-532f-847f-33010935c313)
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, there 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).
If Atossa had desired it, an entire retinue of physicians from Babylonia to Greece would have flocked to her bedside to treat her. Instead, she descended into a fierce and impenetrable loneliness. She wrapped herself in sheets, in a self-imposed quarantine. Darius’ doctors may have tried to treat her, but to no avail. Ultimately, a Greek slave named Democedes persuaded her to allow him to excise the tumor.
Soon after that operation, Atossa mysteriously vanishes from Herodotus’ text. For him, she is merely a minor plot twist. We don’t know whether the tumor recurred, or how or when she died, but the procedure was at least a temporary success. Atossa lived, and she had Democedes to thank for it. And that reprieve from pain and illness whipped her into a frenzy of gratitude and territorial ambition. Darius had been planning a campaign against Scythia, on the eastern border of his empire. Goaded by Democedes, who wanted to return to his native Greece, Atossa pleaded with her husband to turn his campaign westward—to invade Greece. That turn of the Persian empire from east to west, and the series of Greco-Persian wars that followed, would mark one of the definitive moments in the early history of the West. It was Atossa’s tumor, then, that quietly launched a thousand ships. Cancer, even as a clandestine illness, left its fingerprints on the ancient world.


But Herodotus and Imhotep are storytellers, and like all stories, theirs have gaps and inconsistencies. The “cancers” described by them may have been true neoplasms, or perhaps they were hazily describing abscesses, ulcers, warts, or moles. The only incontrovertible cases of cancer in history are those in which the malignant tissue has somehow been preserved. And to encounter one such cancer face-to-face—to actually stare the ancient illness in its eye—one needs to journey to a thousand-year-old gravesite in a remote, sand-swept plain in the southern tip of Peru.
The plain lies at the northern edge of the Atacama Desert, a parched, desolate six-hundred-mile strip caught in the leeward shadow of the giant furl of the Andes that stretches from southern Peru into Chile. Brushed continuously by a warm, desiccating wind, the terrain hasn’t seen rain in recorded history. It is hard to imagine that human life once flourished here, but it did. The plain is strewn with hundreds of graves—small, shallow pits dug out of the clay, then lined carefully with rock. Over the centuries, dogs, storms, and grave robbers have dug out these shallow graves, exhuming history.
The graves contain the mummified remains of members of the Chiribaya tribe. The Chiribaya made no effort to preserve their dead, but the climate is almost providentially perfect for mummification. The clay leaches water and fluids out of the body from below, and the wind dries the tissues from above. The bodies, often placed seated, are thus swiftly frozen in time and space.
In 1990, one such large desiccated gravesite containing about 140 bodies caught the attention of Arthur Aufderheide, a professor at the University of Minnesota in Duluth. Aufderheide is a pathologist by training but his specialty is paleopathology, a study of ancient specimens. His autopsies, unlike Farber’s, are not performed on recently living patients, but on the mummified remains found on archaeological sites. He stores these human specimens in small, sterile milk containers in a vaultlike chamber in Minnesota. There are nearly five thousand pieces of tissue, scores of biopsies, and hundreds of broken skeletons in his closet.
At the Chiribaya site
, Aufderheide rigged up a makeshift dissecting table and performed 140 autopsies over several weeks. One body revealed an extraordinary finding. The mummy was of a young woman in her midthirties, found sitting, with her feet curled up, in a shallow clay grave. When Aufderheide examined her, his fingers found a hard “bulbous mass” in her left upper arm. The papery folds of skin, remarkably preserved, gave way to that mass, which was intact and studded with spicules of bone. This, without question, was a malignant bone tumor, an osteosarcoma, a thousand-year-old cancer preserved inside of a mummy. Aufderheide suspects that the tumor had broken through the skin while she was still alive. Even small osteosarcomas can be unimaginably painful. The woman’s pain, he suggests, must have been blindingly intense.
Aufderheide isn’t the only paleopathologist to have found cancers in mummified specimens. (Bone tumors, because they form hardened and calcified tissue, are vastly more likely to survive over centuries and are best preserved.) “There are other cancers found in mummies where the malignant tissue has been preserved. The oldest of these is an abdominal cancer from Dakhleh in Egypt from about four hundred AD,” he said. In other cases, paleopathologists have not found the actual tumors, but rather signs left by the tumors in the body. Some skeletons were riddled with tiny holes created by cancer in the skull or the shoulder bones, all arising from metastatic skin or breast cancer. In 1914, a team
of archaeologists found a two-thousand-year old Egyptian mummy in the Alexandrian catacombs with a tumor invading the pelvic bone. Louis Leakey,
the anthropologist who dug up some of the earliest known human skeletons, also discovered a jawbone dating from two million years ago from a nearby site that carried the signs of a peculiar form of lymphoma found endemically in southeastern Africa (although the origin of that tumor was never confirmed pathologically). If that finding does represent an ancient mark of malignancy, then cancer, far from being a “modern” disease, is one of the oldest diseases ever seen in a human specimen—quite possibly the oldest.


The most striking finding, though, is not that cancer existed in the distant past, but that it was fleetingly rare. When I asked Aufderheide about this, he laughed. “The early history of cancer,”
he said, “is that there is very little early history of cancer.” The Mesopotamians knew their migraines; the Egyptians had a word for seizures. A leprosy-like illness
, tsara’at, is mentioned in the book of Leviticus. The Hindu Vedas have a medical term for dropsy and a goddess specifically dedicated to smallpox. Tuberculosis was so omnipresent and familiar to the ancients that—as with ice and the Eskimos—distinct words exist for each incarnation of it. But even common cancers, such as breast, lung, and prostate, are conspicuously absent. With a few notable exceptions, in the vast stretch of medical history there is no book or god for cancer.
There are several reasons behind this absence. Cancer is an age-related disease—sometimes exponentially so. The risk of breast cancer
, for instance, is about 1 in 400 for a thirty-year-old woman and increases to 1 in 9 for a seventy-year-old. In most ancient societies, people didn’t live long enough to get cancer. Men and women were long consumed by tuberculosis, dropsy, cholera, smallpox, leprosy, plague, or pneumonia. If cancer existed, it remained submerged under the sea of other illnesses. Indeed, cancer’s emergence in the world is the product of a double negative: it becomes common only when all other killers themselves have been killed. Nineteenth-century doctors often linked cancer to civilization: cancer, they imagined, was caused by the rush and whirl of modern life, which somehow incited pathological growth in the body. The link was correct, but the causality was not: civilization did not cause cancer, but by extending human life spans—civilization unveiled it.
Longevity, although certainly the most important contributor to the prevalence of cancer in the early twentieth century, is probably not the only contributor. Our capacity to detect cancer earlier and earlier, and to attribute deaths accurately to it, has also dramatically increased in the last century. The death of a child with leukemia in the 1850s would have been attributed to an abscess or infection (or, as Bennett would have it, to a “suppuration of blood”). And surgery, biopsy, and autopsy techniques have further sharpened our ability to diagnose cancer. The introduction of mammography to detect breast cancer early in its course sharply increased its incidence—a seemingly paradoxical result that makes perfect sense when we realize that the X-rays allow earlier tumors to be diagnosed.
Finally, changes in the structure of modern life have radically shifted the spectrum of cancers—increasing the incidence of some, decreasing the incidence of others. Stomach cancer, for instance, was highly prevalent in certain populations until the late nineteenth century, likely the result of several carcinogens found in pickling reagents and preservatives and exacerbated by endemic and contagious infection with a bacterium that causes stomach cancer. With the introduction of modern refrigeration (and possibly changes in public hygiene that have diminished the rate of endemic infection), the stomach cancer epidemic seems to have abated. In contrast, lung cancer incidence in men increased dramatically in the 1950s as a result of an increase in cigarette smoking during the early twentieth century. In women, a cohort that began to smoke in the 1950s, lung cancer incidence has yet to reach its peak.
The consequence of these demographic and epidemiological shifts was, and is, enormous. In 1900, as Roswell Park noted, tuberculosis was by far the most common cause of death in America. Behind tuberculosis came pneumonia (William Osler, the famous physician from Johns Hopkins University, called it “captain of the men of death”
), diarrhea, and gastroenteritis. Cancer still lagged
at a distant seventh. By the early 1940s, cancer
had ratcheted its way to second on the list, immediately behind heart disease. In that same span, life expectancy among Americans
had increased by about twenty-six years. The proportion of persons above sixty years—the age when most cancers begin to strike—nearly doubled.
But the rarity of ancient cancers notwithstanding, it is impossible to forget the tumor growing in the bone of Aufderheide’s mummy of a thirty-five-year-old. The woman must have wondered about the insolent gnaw of pain in her bone, and the bulge slowly emerging from her arm. It is hard to look at the tumor and not come away with the feeling that one has encountered a powerful monster in its infancy.
Onkos (#ulink_c0ce9fa0-a28f-5a53-a184-bbd9167aac4a)
Black bile without boiling
causes cancers.
—Galen, AD 130
We have learned nothing
, therefore, about the real cause of cancer or its actual nature. We are where the Greeks were.
—Francis Carter Wood in 1914
It’s bad bile
. It’s bad habits. It’s bad bosses. It’s bad genes.
—Mel Greaves, Cancer: The Evolutionary Legacy, 2000
In some ways disease
does not exist until we have agreed that it does—by perceiving, naming, and responding to it.
—C. E. Rosenberg
Even an ancient monster needs a name. To name an illness is to describe a certain condition of suffering—a literary act before it becomes a medical one. A patient, long before he becomes the subject of medical scrutiny, is, at first, simply a storyteller, a narrator of suffering—a traveler who has visited the kingdom of the ill. To relieve an illness, one must begin, then, by unburdening its story.
The names of ancient illnesses are condensed stories in their own right. Typhus, a stormy disease, with erratic, vaporous fevers, arose from the Greek tuphon, the father of winds—a word that also gives rise to the modern typhoon. Influenza emerged from the Latin influentia because medieval doctors imagined that the cyclical epidemics of flu were influenced by stars and planets revolving toward and away from the earth. Tuberculosis coagulated out of the Latin tuber, referring to the swollen lumps of glands that looked like small vegetables. Lymphatic tuberculosis, TB of the lymph glands, was called scrofula, from the Latin word for “piglet,” evoking the rather morbid image of a chain of swollen glands arranged in a line like a group of suckling pigs.
It was in the time of Hippocrates, around 400 BC, that a word for cancer first appeared in the medical literature: karkinos, from the Greek word for “crab.” The tumor, with its clutch of swollen blood vessels around it, reminded Hippocrates of a crab dug in the sand with its legs spread in a circle. The image was peculiar (few cancers truly resemble crabs), but also vivid. Later writers, both doctors and patients
, added embellishments. For some, the hardened, matted surface of the tumor was reminiscent of the tough carapace of a crab’s body. Others felt a crab moving under the flesh as the disease spread stealthily throughout the body. For yet others, the sudden stab of pain produced by the disease was like being caught in the grip of a crab’s pincers.
Another Greek word would intersect with the history of cancer—onkos, a word used occasionally to describe tumors, from which the discipline of oncology would take its modern name. Onkos was the Greek term for a mass or a load, or more commonly a burden; cancer was imagined as a burden carried by the body. In Greek theater, the same word, onkos, would be used to denote a tragic mask that was often “burdened” with an unwieldy conical weight on its head to denote the psychic load carried by its wearer.
But while these vivid metaphors might resonate with our contemporary understanding of cancer, what Hippocrates called karkinos and the disease that we now know as cancer were, in fact, vastly different creatures. Hippocrates’ karkinos were mostly large, superficial tumors that were easily visible to the eye: cancers of the breast, skin, jaw, neck, and tongue. Even the distinction between malignant and nonmalignant tumors likely escaped Hippocrates: his karkinos included every conceivable form of swelling—nodes, carbuncles, polyps, protrusions, tubercles, pustules, and glands—lumps lumped indiscriminately into the same category of pathology.
The Greeks had no microscopes. They had never imagined an entity called a cell, let alone seen one, and the idea that karkinos was the uncontrolled growth of cells could not possibly have occurred to them. They were, however, preoccupied with fluid mechanics—with waterwheels, pistons, valves, chambers, and sluices—a revolution in hydraulic science originating with irrigation and canal-digging and culminating with Archaemedes discovering his eponymous laws in his bathtub. This preoccupation with hydraulics also flowed into Greek medicine and pathology. To explain illness—all illness—Hippocrates fashioned an elaborate doctrine based on fluids and volumes, which he freely applied to pneumonia, boils, dysentery, and hemorrhoids. The human body, Hippocrates proposed, was composed of four cardinal fluids called humors: blood, black bile, yellow bile, and phlegm. Each of these fluids had a unique color (red, black, yellow, and white), viscosity, and essential character. In the normal body, these four fluids were held in perfect, if somewhat precarious, balance. In illness, this balance was upset by the excess of one fluid.
The physician Claudius Galen, a prolific writer and influential Greek doctor who practiced among the Romans around AD 160, brought Hippocrates’ humoral theory to its apogee. Like Hippocrates, Galen set about classifying all illnesses in terms of excesses of various fluids. Inflammation—a red, hot, painful distension—was attributed to an overabundance of blood. Tubercles, pustules, catarrh, and nodules of lymph—all cool, boggy, and white—were excesses of phlegm. Jaundice was the overflow of yellow bile. For cancer, Galen reserved the most malevolent and disquieting of the four humors: black bile. (Only one other disease, replete with metaphors, would be attributed to an excess of this oily, viscous humor: depression. Indeed, melancholia, the medieval name for “depression,” would draw its name from the Greek melas, “black,” and khole, “bile.” Depression and cancer, the psychic and physical diseases of black bile, were thus intrinsically intertwined.) Galen proposed that cancer was “trapped” black bile—static bile unable to escape from a site and thus congealed into a matted mass. “Of blacke cholor
[bile], without boyling cometh cancer,” Thomas Gale, the English surgeon, wrote of Galen’s theory in the sixteenth century, “and if the humor be sharpe, it maketh ulceration, and for this cause, these tumors are more blacker in color.”
That short, vivid description would have a profound impact on the future of oncology—much broader than Galen (or Gale) may have intended. Cancer, Galenic theory suggested, was the result of a systemic malignant state, an internal overdose of black bile. Tumors were just local outcroppings of a deep-seated bodily dysfunction, an imbalance of physiology that had pervaded the entire corpus. Hippocrates had once abstrusely opined that cancer was “best left untreated,
since patients live longer that way.” Five centuries later, Galen had explained his teacher’s gnomic musings in a fantastical swoop of physiological conjecture. The problem with treating cancer surgically, Galen suggested, was that black bile was everywhere, as inevitable and pervasive as any fluid. You could cut cancer out, but the bile would flow right back, like sap seeping through the limbs of a tree.
Galen died in Rome in 199 AD, but his influence on medicine stretched over the centuries. The black-bile theory of cancer was so metaphorically seductive that it clung on tenaciously in the minds of doctors. The surgical removal of tumors—a local solution to a systemic problem—was thus perceived as a fool’s operation. Generations of surgeons layered their own observations on Galen’s, solidifying the theory even further. “Do not be led away and offer
to operate,” John of Arderne wrote in the mid-1300s. “It will only be a disgrace to you.” Leonard Bertipaglia, perhaps the most influential surgeon of the fifteenth century, added his own admonishment: “Those who pretend
to cure cancer by incising, lifting, and extirpating it only transform a nonulcerous cancer into an ulcerous one. . . . In all my practice, I have never seen a cancer cured by incision, nor known anyone who has.”
Unwittingly, Galen may actually have done the future victims of cancer a favor—at least a temporary one. In the absence of anesthesia and antibiotics, most surgical operations performed in the dank chamber of a medieval clinic—or more typically in the back room of a barbershop with a rusty knife and leather straps for restraints—were disastrous, life-threatening affairs. The sixteenth-century surgeon Ambroise Paré described charring tumors
with a soldering iron heated on coals, or chemically searing them with a paste of sulfuric acid. Even a small nick in the skin, treated thus, could quickly suppurate into a lethal infection. The tumors would often profusely bleed at the slightest provocation.
Lorenz Heister, an eighteenth-century German physician, once described a mastectomy in his clinic as if it were a sacrificial ritual: “Many females can stand the operation
with the greatest courage and without hardly moaning at all. Others, however, make such a clamor that they may dishearten even the most undaunted surgeon and hinder the operation. To perform the operation, the surgeon should be steadfast and not allow himself to become discomforted by the cries of the patient.”
Unsurprisingly, rather than take their chances with such “undaunted” surgeons, most patients chose to hang their fates with Galen and try systemic medicines to purge the black bile. The apothecary
thus soon filled up with an enormous list of remedies for cancer: tincture of lead, extracts of arsenic, boar’s tooth, fox lungs, rasped ivory, hulled castor, ground white-coral, ipecac, senna, and a smattering of purgatives and laxatives. There was alcohol and the tincture of opium for intractable pain. In the seventeenth century, a paste of crab’s eyes, at five shillings a pound, was popular—using fire to treat fire. The ointments and salves grew increasingly bizarre by the century: goat’s dung, frogs, crow’s feet, dog fennel, tortoise liver, the laying of hands, blessed waters, or the compression of the tumor with lead plates.
Despite Galen’s advice, an occasional small tumor was still surgically excised. (Even Galen had reportedly performed such surgeries, possibly for cosmetic or palliative reasons.) But the idea of surgical removal of cancer as a curative treatment was entertained only in the most extreme circumstances. When medicines and operations failed, doctors resorted to the only established treatment for cancer, borrowed from Galen’s teachings: an intricate series of bleeding and purging rituals to squeeze the humors out of the body, as if it were an overfilled, heavy sponge.
Vanishing Humors (#ulink_498a6d01-6dd6-53bb-b481-63e412c92c03)
Rack’t carcasses
make ill Anatomies.
—John Donne
In the winter of 1533, a nineteen-year-old student from Brussels, Andreas Vesalius, arrived at the University of Paris hoping to learn Galenic anatomy and pathology and to start a practice in surgery. To Vesalius’s shock and disappointment, the anatomy lessons at the university were in a preposterous state of disarray. The school lacked a specific space for performing dissections. The basement of the Hospital Dieu, where anatomy demonstrations were held, was a theatrically macabre space where instructors hacked their way through decaying cadavers while dogs gnawed on bones and drippings below. “Aside from the eight muscles
of the abdomen, badly mangled and in the wrong order, no one had ever shown a muscle to me, nor any bone, much less the succession of nerves, veins, and arteries,” Vesalius wrote in a letter. Without a map of human organs to guide them, surgeons were left to hack their way through the body like sailors sent to sea without a map—the blind leading the ill.
Frustrated with these ad hoc dissections, Vesalius decided to create his own anatomical map. He needed his own specimens
, and he began to scour the graveyards around Paris for bones and bodies. At Montfaucon, he stumbled upon the massive gibbet of the city of Paris, where the bodies of petty prisoners were often left dangling. A few miles away, at the Cemetery of the Innocents, the skeletons of victims of the Great Plague lay half-exposed in their graves, eroded down to the bone.
The gibbet and the graveyard—the convenience stores for the medieval anatomist—yielded specimen after specimen for Vesalius, and he compulsively raided them, often returning twice a day to cut pieces dangling from the chains and smuggle them off to his dissection chamber. Anatomy came alive for him in this grisly world of the dead. In 1538, collaborating with artists in Titian’s studio, Vesalius began to publish his detailed drawings in plates and books—elaborate and delicate etchings charting the courses of arteries and veins, mapping nerves and lymph nodes. In some plates, he pulled away layers of tissue, exposing the delicate surgical planes underneath. In another drawing, he sliced through the brain in deft horizontal sections—a human CT scanner, centuries before its time—to demonstrate the relationship between the cisterns and the ventricles.
Vesalius’s anatomical project had started as a purely intellectual exercise but was soon propelled toward a pragmatic need. Galen’s humoral theory of disease—that all diseases were pathological accumulations of the four cardinal fluids—required that patients be bled and purged to squeeze the culprit humors out of the body. But for the bleedings to be successful, they had to be performed at specific sites in the body. If the patient was to be bled prophylactically (that is, to prevent disease), then the purging was to be performed far away from the possible disease site, so that the humors could be diverted from it. But if the patient was being bled therapeutically—to cure an established disease—then the bleeding had to be done from nearby vessels leading into the site.
To clarify this already foggy theory, Galen had borrowed an equally foggy Hippocratic expression, kat' 'i'xiu—Greek for “straight into”—to describe isolating the vessels that led “straight into” tumors. But Galen’s terminology had pitched physicians into further confusion. What on earth, they wondered, had Galen meant by “straight into”? Which vessels led “straight into” a tumor or an organ, and which led the way out? The instructions became a maze of misunderstanding. In the absence of a systematic anatomical map—without the establishment of normality—abnormal anatomy was impossible to fathom.
Vesalius decided to solve the problem by systematically sketching out every blood vessel and nerve in the body, producing an anatomical atlas for surgeons. “In the course of explaining the opinion
of the divine Hippocrates and Galen,” he wrote in a letter, “I happened to delineate the veins on a chart, thinking that thus I might be able easily to demonstrate what Hippocrates understood by the expression kat' 'i'xiu, for you know how much dissension and controversy on venesection was stirred up, even among the learned.”
But having started this project, Vesalius found that he could not stop. “My drawing of the veins pleased the professors of medicine and all the students so much that they earnestly sought from me a diagram of the arteries and also one of the nerves. . . . I could not disappoint them.” The body was endlessly interconnected: veins ran parallel to nerves, the nerves were connected to the spinal cord, the cord to the brain, and so forth. Anatomy could only be captured in its totality, and soon the project became so gargantuan and complex that it had to be outsourced to yet other illustrators to complete.
But no matter how diligently Vesalius pored through the body, he could not find Galen’s black bile. The word autopsy comes from the Greek “to see for oneself”; as Vesalius learned to see for himself, he could no longer force Galen’s mystical visions to fit his own. The lymphatic system carried a pale, watery fluid; the blood vessels were filled, as expected, with blood. Yellow bile was in the liver. But black bile—Galen’s oozing carrier of cancer and depression—could not be found anywhere.
Vesalius now found himself in a strange position. He had emerged from a tradition steeped in Galenic scholarship; he had studied, edited, and republished Galen’s books. But black bile—that glistening centerpiece of Galen’s physiology—was nowhere to be found. Vesalius hedged about his discovery. Guiltily, he heaped even more praise on the long-dead Galen. But, an empiricist to the core, Vesalius left his drawings just as he saw things, leaving others to draw their own conclusions. There was no black bile. Vesalius had started his anatomical project to save Galen’s theory, but, in the end, he quietly buried it.


In 1793, Matthew Baillie, an anatomist in London, published a textbook called The Morbid Anatomy of Some of the Most Important Parts of the Human Body. Baillie’s book, written for surgeons and anatomists, was the obverse of Vesalius’s project: if Vesalius had mapped out “normal” anatomy, Baillie mapped the body in its diseased, abnormal state. It was Vesalius’s study read through an inverted lens. Galen’s fantastical speculations about illnesses were even more at stake here. Black bile may not have existed discernably in normal tissue, but tumors should have been chock-full of it. But none was to be found. Baillie described cancers of the lung (“as large as an orange”
), stomach (“a fungous appearance”
), and the testicles (“a foul deep ulcer”
) and provided vivid engravings of these tumors. But he could not find the channels of bile anywhere—not even in his orange-size tumors, nor in the deepest cavities of his “foul deep ulcers.” If Galen’s web of invisible fluids existed, then it existed outside tumors, outside the pathological world, outside the boundaries of normal anatomical inquiry—in short, outside medical science. Like Vesalius, Baillie drew anatomy and cancer the way he actually saw it. At long last, the vivid channels of black bile, the humors in the tumors, that had so gripped the minds of doctors and patients for centuries, vanished from the picture.
“Remote Sympathy” (#ulink_5572c14a-6ce8-5415-8620-c204a488cca3)
In treating of cancer
, we shall remark, that little or no confidence should be placed either in internal . . . remedies, and that there is nothing, except the total separation of the part affected.
—A Dictionary of Practical Surgery, 1836
Matthew Baillie’s Morbid Anatomy laid the intellectual foundation for the surgical extractions of tumors. If black bile did not exist, as Baillie had discovered, then removing cancer surgically might indeed rid the body of the disease. But surgery, as a discipline, was not yet ready for such operations. In the 1760s, a Scottish surgeon, John Hunter, Baillie’s maternal uncle, had started to remove tumors from his patients in a clinic in London in quiet defiance of Galen’s teachings. But Hunter’s elaborate studies—initially performed on animals and cadavers in a shadowy menagerie in his own house—were stuck at a critical bottleneck. He could nimbly reach down into the tumors and, if they were “movable” (as he called superficial, noninvasive cancers), pull them out without disturbing the tender architecture of tissues underneath. “If a tumor is not only movable
but the part naturally so,” Hunter wrote, “they may be safely removed also. But it requires great caution to know if any of these consequent tumors are within proper reach, for we are apt to be deceived.”
That last sentence was crucial. Albeit crudely, Hunter had begun to classify tumors into “stages.” Movable tumors were typically early-stage, local cancers. Immovable tumors were advanced, invasive, and even metastatic. Hunter concluded that only movable cancers were worth removing surgically. For more advanced forms of cancer, he advised an honest, if chilling, remedy reminiscent of Imhotep’s: “remote sympathy.”*
Hunter was an immaculate anatomist, but his surgical mind was far ahead of his hand. A reckless and restless man with nearly maniacal energy who slept only four hours a night, Hunter had practiced his surgical skills endlessly on cadavers from every nook of the animal kingdom—on monkeys, sharks, walruses, pheasants, bears, and ducks. But with live human patients, he found himself at a standstill. Even if he worked at breakneck speed, having drugged his patient with alcohol and opium to near oblivion, the leap from cool, bloodless corpses to live patients was fraught with danger. As if the pain during surgery were not bad enough, the threat of infections after surgery loomed. Those who survived the terrifying crucible of the operating table often died even more miserable deaths in their own beds soon afterward.


In the brief span between 1846 and 1867, two discoveries swept away these two quandaries that had haunted surgery, thus allowing cancer surgeons to revisit the bold procedures that Hunter had tried to perfect in London.
The first of these discoveries, anesthesia, was publicly demonstrated in 1846 in a packed surgical amphitheater at Massachusetts General Hospital, less than ten miles from where Sidney Farber’s basement laboratory would be located a century later. At about ten o’clock on the morning of October 16, a group of doctors gathered in a pitlike room at the center of the hospital. A Boston dentist, William Morton, unveiled a small glass vaporizer, containing about a quart of ether, fitted with an inhaler. He opened the nozzle and asked the patient, Edward Abbott, a printer, to take a few whiffs of the vapor. As Abbott lolled into a deep sleep, a surgeon stepped into the center of the amphitheater and, with a few brisk strokes, deftly made a small incision in Abbott’s neck and closed a swollen, malformed blood vessel (referred to as a “tumor,” conflating malignant and benign swellings) with a quick stitch. When Abbott awoke a few minutes later, he said, “I did not experience pain
at any time, though I knew that the operation was proceeding.”
Anesthesia—the dissociation of pain from surgery—allowed surgeons to perform prolonged operations, often lasting several hours. But the hurdle of postsurgical infection remained. Until the mid-nineteenth century, such infections were common and universally lethal, but their cause remained a mystery. “It must be some subtle principle
contained [in the wound],” one surgeon concluded in 1819, “which eludes the sight.”
In 1865, a Scottish surgeon named Joseph Lister made an unusual conjecture on how to neutralize that “subtle principle” lurking elusively in the wound. Lister began with an old clinical observation: wounds left open to the air would quickly turn gangrenous, while closed wounds would often remain clean and uninfected. In the postsurgical wards of the Glasgow infirmary, Lister had again and again seen an angry red margin begin to spread out from the wound and then the skin seemed to rot from inside out, often followed by fever, pus, and a swift death (a bona fide “suppuration”).
Lister thought of a distant, seemingly unrelated experiment. In Paris, Louis Pasteur, the great French chemist, had shown that meat broth left exposed to the air would soon turn turbid and begin to ferment, while meat broth sealed in a sterilized vacuum jar would remain clear. Based on these observations, Pasteur had made a bold claim: the turbidity was caused by the growth of invisible microorganisms—bacteria—that had fallen out of the air into the broth. Lister took Pasteur’s reasoning further. An open wound—a mixture of clotted blood and denuded flesh—was, after all, a human variant of Pasteur’s meat broth, a natural petri dish for bacterial growth. Could the bacteria that had dropped into Pasteur’s cultures in France also be dropping out of the air into Lister’s patients’ wounds in Scotland?
Lister then made another inspired leap of logic. If postsurgical infections were being caused by bacteria, then perhaps an antibacterial process or chemical could curb these infections. It “occurred to me,”
he wrote in his clinical notes, “that the decomposition in the injured part might be avoided without excluding the air, by applying as a dressing some material capable of destroying the life of the floating particles.”
In the neighboring town of Carlisle, Lister had observed sewage disposers cleanse their waste with a cheap, sweet-smelling liquid containing carbolic acid. Lister began to apply carbolic acid paste to wounds after surgery. (That he was applying a sewage cleanser to his patients appears not to have struck him as even the slightest bit unusual.)
In August 1867, a thirteen-year-old
boy who had severely cut his arm while operating a machine at a fair in Glasgow was admitted to Lister’s infirmary. The boy’s wound was open and smeared with grime—a setup for gangrene. But rather than amputating the arm, Lister tried a salve of carbolic acid, hoping to keep the arm alive and uninfected. The wound teetered on the edge of a terrifying infection, threatening to become an abscess. But Lister persisted, intensifying his application of carbolic acid paste. For a few weeks, the whole effort seemed hopeless. But then, like a fire running to the end of a rope, the wound began to dry up. A month later, when the poultices were removed, the skin had completely healed underneath.
It was not long before Lister’s invention was joined to the advancing front of cancer surgery. In 1869, Lister removed a breast tumor
from his sister, Isabella Pim, using a dining table as his operating table, ether for anesthesia, and carbolic acid as his antiseptic. She survived without an infection (although she would eventually die of liver metastasis three years later). A few months later, Lister performed an extensive amputation
on another patient with cancer, likely a sarcoma in a thigh. By the mid-1870s, Lister was routinely operating on breast cancer and had extended his surgery to the cancer-afflicted lymph nodes under the breast.


Antisepsis and anesthesia were twin technological breakthroughs that released surgery from its constraining medieval chrysalis. Armed with ether and carbolic soap, a new generation of surgeons lunged toward the forbiddingly complex anatomical procedures that Hunter and his colleagues had once concocted on cadavers. An incandescent century of cancer surgery emerged; between 1850 to 1950, surgeons brazenly attacked cancer by cutting open the body and removing tumors.
Emblematic of this era was the prolific Viennese surgeon Theodor Billroth. Born in 1821, Billroth studied music and surgery with almost equal verve. (The professions still often go hand in hand. Both push manual skill to its limit; both mature with practice and age; both depend on immediacy, precision, and opposable thumbs.) In 1867, as a professor in Berlin, Billroth launched a systematic study of methods to open the human abdomen to remove malignant masses. Until Billroth’s
time, the mortality following abdominal surgery had been forbidding. Billroth’s approach to the problem was meticulous and formal: for nearly a decade, he spent surgery after surgery simply opening and closing abdomens of animals and human cadavers, defining clear and safe routes to the inside. By the early 1880s, he had established the routes: “The course so far is already
sufficient proof that the operation is possible,” he wrote. “Our next care, and the subject of our next studies, must be to determine the indications, and to develop the technique to suit all kinds of cases. I hope we have taken another good step forward towards securing unfortunate people hitherto regarded as incurable.”
At the Allgemeines Krankenhaus, the teaching hospital in Vienna where he was appointed a professor, Billroth and his students now began to master and use a variety of techniques to remove tumors from the stomach, colon, ovaries, and esophagus, hoping to cure the body of cancer. The switch from exploration to cure produced an unanticipated challenge. A cancer surgeon’s task was to remove malignant tissue while leaving normal tissues and organs intact. But this task, Billroth soon discovered, demanded a nearly godlike creative spirit.
Since the time of Vesalius, surgery had been immersed in the study of natural anatomy. But cancer so often disobeyed and distorted natural anatomical boundaries that unnatural boundaries had to be invented to constrain it. To remove the distal end of a stomach filled with cancer, for instance, Billroth had to hook up the pouch remaining after surgery to a nearby piece of the small intestine. To remove the entire bottom half of the stomach, he had to attach the remainder to a piece of distant jejunum. By the mid-1890s, Billroth had operated on forty-one patients with gastric carcinoma using these novel anatomical reconfigurations. Nineteen of these patients had survived the surgery.
These procedures represented pivotal advances in the treatment of cancer. By the early twentieth century, many locally restricted cancers (i.e., primary tumors without metastatic lesions) could be removed by surgery. These included uterine and ovarian cancer, breast and prostate cancer, colon cancer, and lung cancer. If these tumors were removed before they had invaded other organs, these operations produced cures in a significant fraction of patients.
But despite these remarkable advances, some cancers—even seemingly locally restricted ones—still relapsed after surgery, prompting second and often third attempts to resect tumors. Surgeons returned to the operating table
and cut and cut again, as if caught in a cat-and-mouse game, as cancer was slowly excavated out of the human body piece by piece.
But what if the whole of cancer could be uprooted at its earliest stage using the most definitive surgery conceivable? What if cancer, incurable by means of conventional local surgery, could be cured by a radical, aggressive operation that would dig out its roots so completely, so exhaustively, that no possible trace was left behind? In an era captivated by the potency and creativity of surgeons, the idea of a surgeon’s knife extracting cancer by its roots was imbued with promise and wonder. It would land on the already brittle and combustible world of oncology like a firecracker thrown into gunpowder.

Конец ознакомительного фрагмента.
Текст предоставлен ООО «ЛитРес».
Прочитайте эту книгу целиком, купив полную легальную версию (https://www.litres.ru/siddhartha-mukherjee/the-emperor-of-all-maladies/) на ЛитРес.
Безопасно оплатить книгу можно банковской картой Visa, MasterCard, Maestro, со счета мобильного телефона, с платежного терминала, в салоне МТС или Связной, через PayPal, WebMoney, Яндекс.Деньги, QIWI Кошелек, бонусными картами или другим удобным Вам способом.