History of breast cancer treatments

I need a paper not too long about 6 pages only, I have started but it needs more development as the professor ask for it. I need to provide a thesis statement and the paper need to be documented properly MLA style I have turtinent it 

1

Don't use plagiarized sources. Get Your Custom Essay on
History of breast cancer treatments
Just from $13/Page
Order Essay

The story of breast cancer is told in the acts and arti-
facts of the human struggle against disease. It is an
epic tale that follows the concepts of illness from the
work of evil spirits or of offended gods to the results
of identifiable physical causes, and the healing arts
from mysticism to the tools of modern science. The
following is a brief history of breast cancer in the
Western world.

PREHISTORY AND THE ANCIENT WORLD

Prior to recorded history, life was undoubtedly short,
and as cancer is predominantly a disease of maturity
one suspects that cancer was a poor competitor
among causes of mortality. The study of primitive
peoples indicates that for the ill, rituals, potions, and
recipes at the hands of magicians, witch doctors, and
folk healers were the usual recourses. In ancient
Babylon (2100–689 BC) it was common practice to
place the ailing in public places for the recommen-
dations of passersby, but professional healers were
also recognized. The Code of Hammurabi, inscribed
on a pillar in Babylon, indicated that healers were
paid fees for their services and were penalized for
surgical deaths with amputation of their hands.1

Before the third millennium BC, physicians had
learned the futility of treating certain tumors of the
breast. Among the eight extant Egyptian medical
papyri, The Edwin Smith Surgical Papyrus is
believed to contain the first reference to breast can-
cer (Figure 1–1). This surgical text, penned in hier-
atic script, is the incomplete and fragmented copy of
an original document that probably dates back to the
pyramid age of Egypt (3000–2500 BC) and was pos-

sibly written by Imhotep, the physician-architect who
practiced medicine and designed the step pyramid in
Egypt in the 30th century BC.2 It provides the earli-
est references to suturing of wounds and to cauteri-
zation with fire drills. More pertinently, it includes
the diagnosis and treatment of eight cases of ailments
of the “breast,” meaning of the bones and soft tissues

1

History of Breast Cancer
WILLIAM L. DONEGAN

Figure 1–1. Column VIII of The Edwin Smith Surgical Papyrus, a
copy of the first document believed to describe cancer of the
breast, circa 3000 BC. Used with permission from The Classics of
Surgery Library.2

2

  • BREAST CANCER
  • of the anterior thorax, all in men and most due to
    injuries. One of the five cases relating to soft tissues
    (Case 45) describes “bulging tumors” in the breast.
    The author writes that if the tumors have spread over
    the breast, are cool to the touch, and are bulging,
    there is no treatment. Whether this case was a rare
    cancer of the male breast is conjectural, but in stark
    contrast to the physician’s active recommendations
    for the other cases, he recognized this one as sinister;
    and his conviction that no treatment would help
    appears to have been based on established practice.

    GREEK AND ROMAN PERIOD
    (460 BC–475 AD)

    Ancient Greece was pervaded by a rich mythology
    based on a belief in close associations between
    humans and gods. Historians speculate that the god
    of medicine, Aesculapius, may have had origin in a
    physician who lived around the time of the siege
    of Troy (≈1300 BC) and to whom were attributed
    miracles of healing. In the Iliad, Homer mentioned
    Aesculapius’ two sons as “good physicians” who
    had come to join the siege.3 On the seal of the Amer-
    ican College of Surgeons, Aesculapius is pictured
    seated, holding his staff entwined with a serpent, the
    symbol of life and wisdom. Early Greeks sought
    cures by sleeping in the abaton at the temples of
    Aesculapius and enjoying the associated baths and
    recreations, forerunners of modern health spas.
    Votive offerings in the form of breasts found at such
    sites offer evidence that some came hoping for cure
    of breast disease (Figure 1–2).

    Greek medicine and surgery became the most
    sophisticated of its time. In the course of his con-
    quests, Alexander the Great of Macedonia (356–323
    BC) founded the city of Alexandria on the Nile delta
    in 332 BC, and a famous medical school arose there
    around 300 BC. The library at Alexandria was the
    largest of its time, housing more than 700,000 scrolls.
    Many prominent Greek and Roman physicians stud-
    ied, taught, and practiced in Alexandria. The study of
    anatomy was based on dissection of human bodies
    and surgery flourished; vascular ligatures were used.

    Physicians of the Hellenistic period provide
    vivid accounts of breast cancer. The Greek term
    “karkinoma” was used to describe malignant

    growths and “scirrhous” to describe particularly
    hard, solid tumors. “Cacoethes” referred to an early
    or a probable malignancy. A “hidden” cancer was
    one not ulcerating the skin. In an anecdote,
    Herodotus (484–425 BC), historian of the wars
    between Greece and Persia, claimed that Demo-
    cedes, a Persian physician living in Greece, cured
    the wife of Persian King Darius of a breast tumor
    that had ulcerated and spread.

    Hippocrates (460–375 BC), whose legacy, the
    Corpus Hippocraticum, may have been the work of
    more than one person, was the most prominent of
    Greek physicians. He maintained that every disease
    was distinctive and arose from natural causes, not
    from gods or spirits.4 He also believed in the power of
    nature to heal and in a humoral origin of disease. In
    his view, a balance of the four bodily fluids, blood,
    phlegm, yellow bile, and black bile (later linked to
    sanguine, phlegmatic, choleric, and melancholy dis-
    positions by Galen) was necessary for good health.
    Hippocrates described cases of breast cancer in detail.
    One of his case histories was of a woman of Abdera
    who had a carcinoma of the breast with bloody dis-
    charge from her nipple. Attaching a beneficial effect
    to the bleeding, he noted that when the discharge
    stopped, she died. Similarly, Hippocrates associated
    cessation of menstrual bleeding with breast cancer
    and sought to restore menstruation in young sufferers.
    His detailed description of the inexorable course of
    advancing breast cancer rings true today. He said that

    Figure 1–2. Votive offerings from an Etruscan temple include a
    vagina, a uterus, an ear, an eye, and a breast (lower central). Repro-
    duced with permission from Lyons AS and Petrucelli RJ.6

    History of Breast Cancer 3

    hard tumors appear in the breast, become increasingly
    firm, contain no pus, and spread to other parts of the
    body. As the disease progresses, the patient develops
    bitter taste, refuses food, develops pain that shoots
    from the breast to the neck and shoulder blades, com-
    plains of thirst, and becomes emaciated. From this
    point death was certain. He advised no treatment for
    hidden breast cancers because treatment was futile
    and shortened the patient’s life.

    In the ascendant Roman Empire, physicians were
    guided largely by Greek medicine. Around 30 AD, the
    Roman physician Aulus Cornelius Celsus (42 BC–37
    AD) noted that the breasts of women were frequent
    sites of cancer. Celsus described breast cancer in his
    manuscript, De Medicina, and defined four stages.
    The first was cacoethes, followed by carcinoma with-
    out skin ulceration, carcinoma with ulceration, and,
    finally, “thymium,” an advanced exophytic and some-
    times bleeding lesion, the appearance of which sug-
    gested to him the flowers of thyme. Celsus recom-
    mended excision for the cacoethes but no treatment
    for other stages. In situations of uncertainty, the tumor
    was treated first with caustics, and if the symptoms
    improved, it was a cacoethes; if they worsened, it was
    a carcinoma. Some masses for which treatment was
    successful might have been fibroadenomas, phyl-
    lodes tumors, or even tuberculosis.

    Leonides, a surgeon of the Alexandrian school,
    described surgical removal of breast cancers during
    this time.4 Leonides said that with the patient supine
    he cut into the sound part of the breast and used a
    technique of alternately cutting and cauterizing with
    hot irons to control bleeding. The resection was car-
    ried through normal tissues wide of the tumor and
    customized to the extent of involvement. The opera-
    tion was concluded with a general cauterization to
    destroy any residual disease. Poultices were then
    applied to the wound to promote healing. He
    explained that excision was used selectively for
    tumors in the upper part of the breast of limited
    extent, and he specifically advised against surgery if
    the whole breast was hardened or if the tumor was
    fixed to the chest wall. Leonides was perhaps the
    first to record that breast cancers spread to the
    axilla. Complete and thorough excision of breast
    malignancies has been a cardinal principle of
    surgery since the time of Leonides.

    The teachings of the Greek physician, Galen of
    Pergamum (129–200 BC), on the subject of breast
    cancer reached far beyond his time. Born of a
    wealthy and educated family in Asia Minor, he
    traveled and studied widely. Galen became sur-
    geon to gladiators in Pergamum and finally prac-
    ticed in Rome, attending the emperor Marcus
    Aurelius. His vast experience, clinical acumen,
    investigative approach to knowledge, and prolific,
    authoritative writings (400 treatises) gained Galen
    enormous respect. For the next 1,500 years,
    Galen’s teachings guided medical practice, and his
    animal dissections provided the bases for human
    anatomy and physiology.

    Galen revered Hippocrates and adopted his
    humoral theory of disease. In Galen’s view, breast
    cancer was a systemic disease caused by an excess of
    black bile in the blood (ie, melancholia). Black bile
    was formed in the liver from blood elements and
    absorbed in the spleen; malfunction of either of these
    organs caused an excess of black bile, which thick-
    ened the blood, and where black bile accumulated,
    carcinoma developed as hard, non-tender tumors that
    ulcerated if the bile was particularly acrid. Like Hip-
    pocrates, he noted that carcinomas were predisposed
    to accumulate in the breasts of women who had
    ceased to menstruate, a recurring theme and doubt-
    less a reference to the frequency of cancer in post-
    menopausal women. This observation supported
    Galen’s belief that menstruation, and the practice of
    bleeding, served to clear the body of excess black
    bile. He likened the dilated veins that radiated from
    carcinomas to the legs of a crab; as a result, the crab
    became a symbol for cancer. Leonides had also
    likened cancers to crabs, but rather because the tena-
    cious adherence to surrounding tissues mimicked the
    crab’s pinchers. For early cancers, Galen recom-
    mended purging, bleeding, diet, and topicals. Ulcer-
    ating cancers were treated with caustics or cleansed
    and treated with zinc oxide.

    In operating for breast cancer, Galen’s approach
    was less modern than that of Leonides before him.
    Galen condemned the use of ligatures, and although
    he was aware of the dangers of excessive blood loss,
    he preferred to let the blood run unchecked and to
    express the dark, dilated veins in order to rid them of
    the morbid black bile. The cancer was removed at

    4 BREAST CANCER

    the boundary between diseased and healthy parts,
    sparing the cautery out of concern for destroying too
    much tissue. After Galen, medicine languished into
    a contented observance of his teachings, and the
    Middle Ages intervened, temporarily halting further
    medical progress.

    MIDDLE AGES (476–1500 AD)

    The Middle Ages, a period of roughly 1,000 years,
    began with the collapse of the Roman Empire in 476
    and ended with the Renaissance and discovery of the
    New World in 1492. With the Middle Ages came
    feudalism, bubonic plague, crusades, and the age of
    faith. Papal influence spread in the form of the Holy
    Roman Empire, and human dissection was prohib-
    ited by Papal decree; opposition to church doctrine
    constituted heresy. To save his soul, the astronomer
    Copernicus (1473–1543 AD) was forced to rescind
    his thesis that the earth circled the sun rather than
    the reverse, and the physician Michael Servetus
    (1511–1553), discoverer of the pulmonary circula-
    tion, was burned alive for heresy. Meanwhile,
    monastic scribes in Christian Europe quietly pre-
    served medical knowledge, principally that of
    Galen, by copying and illuminating surviving
    ancient manuscripts, manuscripts that were in little
    demand during an era of widespread illiteracy.
    Monks dispensed folk remedies, and surgery was
    discouraged. Amputation of the breast was depicted
    by the church as a form of torture in the story of
    St. Agatha, the patron saint of breast disease5 (Fig-
    ure 1–3). Many miraculous cures were attributed to
    saints. Faith healing by the laying on of hands was
    among the remedies, a practice that endured to
    recent times. Folk medicine included application of
    fresh bisected puppies and cats.

    After the death of the prophet Muhammad
    (570–632 AD), the rise of Islam resulted in the Arab
    conquest of the southern shores of the Mediterranean
    from Persia to Spain, bringing to an end the medical
    center in Alexandria. Medical documents that sur-
    vived were translated into Arabic for study and pre-
    served; translated later from Arabic into Latin, the
    language of medicine in Europe, they re-entered the
    continent. In addition to preserving the past, Arabic
    medicine was noted for expertise in pharmacy and

    for establishing fine hospitals. Among the most
    influential physicians of this period were Avicenna
    (980–1037 AD), the Jewish physician Maimonides
    (1135–1204 AD) and Albucasis (936–1013).6 Avi-
    cenna’s reputation rivaled that of Galen, but he had
    no new insights about breast cancer. Albucasis in
    Moorish Spain favored the cautery and caustic appli-
    cations for treatment of breast cancer but admitted
    that he had never cured a case of breast cancer and
    knew of no one who had. Caustic paste (a mixture of
    zinc chloride, stibnite, and Sanguinaria canadensis)
    was used for treatment of breast cancer in the United
    States as late as the 1950s.7 The paste was applied to
    the involved breast to cause progressive tissue necro-
    sis, which was then cut away or allowed to slough
    and to heal by granulation. Continued use of charms,
    prayers, medicaments, and caustics in conjunction
    with surgery and modern methods is a reminder that
    treatments for breast cancer progressed through his-
    tory not by substitution, but by addition.

    Figure 1–3. Saint Agatha, the patron saint of breast disease, was
    martyred for her Christian beliefs. Her torture included amputation of
    the breasts shown here in a painting by Anthony Van Dyck.5

    History of Breast Cancer 5

    In the late Middle Ages, Henri de Mondeville
    (1260–1320 AD), surgeon to the king of France,
    refined Galen’s black bile theory with a distinction
    between black bile from the liver, which caused a
    hard tumor in the breast (a sclerosis), and twice
    combusted black bile derived from breakdown of the
    other three other body humors, which caused a true
    cancer. He described true breast cancer as ulcerated
    with thick margins and having an offensive odor.
    The treatment: diet and purging, with operation only
    if the cancer could be completely excised; de Mon-
    deville appreciated that incomplete removal often
    resulted in a non-healing wound.8

    RENAISSANCE (SIXTEENTH
    TO EIGHTEENTH CENTURIES)

    The Middle Ages ended with the Renaissance. This
    period of approximately 200 years, also known as
    The Enlightenment, saw a rejection of medieval val-
    ues and a rebirth of interest in secular art, in science,
    and in exploration of the world and the human body.
    With the Renaissance came badly needed formal
    training for physicians. The University of Salerno,
    founded around 1200 AD, was the first organized
    medical school in Europe. Free of clerical influence
    and progressive for its time, Salerno served as the
    precursor of prominent schools of medicine in
    France, England, and elsewhere on the continent.
    The Royal College of Physicians was established in
    London in 1518, and the first medical journal, the
    Ephemerides, appeared in 1670.

    Surgeons became more respectable. Tradition-
    ally unlettered craftsmen whose operations were
    directed by physicians, surgeons became indepen-
    dent practitioners. Incorporated as barber-surgeons
    in England since 1461, surgeons were officially
    separated from barber guilds in 1745. The French
    Academie de Chirurgie, established in 1731, pro-
    duced the first journal for surgeons, Memoires,
    which in 1757 published Henri LeDran’s thesis that
    breast cancer had a local origin, providing an impe-
    tus for surgical cure.9

    The Renaissance in medicine brought a critical
    reexamination of anatomy and physiology and a
    decline of Galen’s authority. Publication of
    Andreas Vesalius’s De Humani Corporis Fabrica in

    1543 marked the beginning. This volume of
    anatomical drawings, based on the young professor
    of surgery at Padua’s own dissections of human
    cadavers, illustrated the errors of Galen’s anatomy
    and stimulated further interest in human anatomy.10

    The Fabrica provided no useful details of the
    female breast. However, 300 years later, Sir Astley
    P. Cooper (1768–1841), surgeon to Guy’s Hospital
    in London, illustrated with desiccated specimens
    the suspensory ligaments of the breast that bear his
    name. The Parisian anatomist Marie-Philibert-Con-
    stant Sappey (1810–1896) illustrated the lymphat-
    ics of the breast, a name that endures as Sappey’s
    subareolar plexus.11,12

    Each anatomic discovery generated new theories
    about breast cancer, but to little advantage. John
    Hunter (1728–1793), the father of investigative
    surgery, conceived that coagulation of lymph rather
    than black bile was responsible for carcinoma of the
    breast and the associated cancerous nodes. Boer-
    haave of Leyden (1668–1738) postulated that neural
    fluid “liquor nervorum” might be the instigator of
    breast cancer, whereas others believed that inspis-
    sated milk within the mammary ducts generated
    cancers. Trauma to the breast was believed to cause
    leakage into the tissues, which created irritation,
    induration, and malignant change. Observing the
    rapid growth of ulcerating breast cancers, Claude-
    Nicholas le Cat (1700–1768) in Rouen postulated
    that exposure to air was a stimulant to cancers, a
    tenacious idea persisting in some laity today. Anec-
    dotes of multiple-affected family members sup-
    ported the suspicion that breast cancer was infec-
    tious long before the hereditary aspect of the disease
    became known in the twentieth century. The deadly
    spread of malignancy was attributed to circulating
    humors or to a general diathesis. The suspicion of a
    “cancer prone” personality lingers but remains
    unconfirmed by modern psychological research.13

    Breast lumps continued to fuel controversy
    about the nature of a “schirrous,” the hard tumor that
    generated concern for patient and physician.
    Whether schirrous was benign, a stage of cancer, or
    a precursor that became cancer by a process of
    “acrimony” remained in doubt. Observation or
    immediate treatment divided opinions. Opinions on
    the worth of surgery varied. Extended survival of

    6 BREAST CANCER

    occasional untreated cases, coupled with the consid-
    erable risk and poor results of mastectomy, sup-
    ported a nihilistic attitude among many physicians.
    Others shared the opinion of Nicolaes Tulp
    (1593–1674) of Amsterdam, who saw the need for
    early surgery. “The sole remedy is a timely operation,”
    he said.4 For the most part, the fearsome prospect of
    an operation was delayed until bulky growth, pain,
    or ulceration made obvious both the diagnosis and
    the need. Informed surgeons recognized tumor attach-
    ment to the chest wall, sternal pain due to deep inva-
    sion or involvement of the internal mammary nodes
    (described by Petrus Camper in 1777), poor general
    health, or a diathesis-revealing “melancholy” appear-
    ance as contraindications to mastectomy.

    Without anesthesia or antisepsis, mastectomies
    were a painful and dangerous ordeal customarily car-
    ried out in the patient’s home. The procedure varied
    from impalement of the breast with needles and
    ropes for traction followed by swift amputation
    through the base, leaving a large open wound as illus-
    trated by Johann Scultetus (1595–1645) in his Arma-
    mentarium Chirurgicum, to the alternative of incis-
    ing the skin and enucleating the tumor by hand.4 The
    prevailing opinion was to leave the wound open to
    minimize the risk of infection. From 2 to 10 minutes
    were required for the operation, depending on the
    technique. Ligatures, if used, were led out through
    the wound to be withdrawn later, after necrosis or
    infection loosened them. Painful re-explorations of
    the wound on subsequent days were performed to
    inspect for infection or to remove additional tumor;
    the major threats were secondary hemorrhage or
    potentially fatal infection. In various illustrations, the
    patient’s hands were tied behind her back or assis-
    tants restrained her while another assistant caught
    jets of blood in a pan. A cauterizing iron provided
    hemostasis, and steam issued from the wound where
    it seared the flesh. The company included a dour,
    attending physician and often an anguished family
    standing in witness. Students of breast cancer should
    not miss the touching account of such an operation in
    Scotland told by John Brown.14 The rigors of surgery
    were such that alternative treatment with compres-
    sion of the breast using metal plates or strapping, not
    entirely devoid of pain and occasional necrosis, con-
    tinued to survive into the nineteenth century.

    Expert surgeons operating in major centers dur-
    ing these times enlarged mastectomies to include all
    morbid parts. In Paris, Jean Louis Petit (1674–1750)
    removed both the breast and diseased nodes in his
    operations, and in 1774, Bernhard Perilhe reported
    removing the pectoralis major muscle as well. A
    healed wound was the customary end point for
    declaring a surgeon’s success; few bothered with
    further follow-up. In a report by Richard Wiseman
    (1622–1676), surgeon to Charles II, among twelve
    mastectomies, two patients (17%) died from the
    operation, eight died shortly afterwards from pro-
    gressive cancer, and two of the 12 were declared
    “cured” for undisclosed lengths of time.4

    NINETEENTH CENTURY

    From the oncologic standpoint, the nineteenth cen-
    tury was truly a giant step forward. Major advances
    were made in human pathology and in the safety of
    surgery. Hand washing was promoted by the Hun-
    garian physician Ignac Semmelweis (1818–1865)
    and by Oliver Wendell Holmes, MD (1809–1894),
    Professor of Anatomy and Physiology at Harvard
    University. Building on Louis Pasteur’s (1822–1895)
    discovery of “putrefying” bacteria, Joseph Lister
    (1827–1912) in Glasgow introduced surgical anti-
    sepsis with carbolic acid spray in 1867.15 Adoption
    of aseptic techniques (ie, steam sterilization) first by
    Ernst von Bergmann of Berlin in 1886, the surgical
    mask by the Pole Johannes von Mikuliez-Radecki in
    1886, and sterile rubber surgical gloves by William
    S. Halsted in 1890 further reduced contamination.16

    Successful demonstration of general anesthesia by
    William T. Morton in Boston in 1846 allowed
    unprecedented development of surgery; operations
    became more acceptable, and for the first time sur-
    geons could concentrate on precision rather than
    haste. Blood transfusions became safe after 1900
    when Karl Landsteiner in Austria discovered blood
    groups. All of the current technology for treatment
    of breast cancer had their beginnings in this century;
    only chemotherapy remained for development in the
    years to come.

    The microscope was the key to progress in
    pathology. Building on Anton van Leewenhoek’s
    (1674–1723) work with lenses, perfection of the com-

    History of Breast Cancer 7

    pound achromatic microscope in Germany opened
    the world of microscopic anatomy, and Germany was
    the center of this new science under the leadership of
    Johannes Müller at the University of Berlin.

    Early in the century, the microscopic work of
    Matthias Schleiden (1804–1881), a botanist at the
    University of Jena, and of Theodor Schwann
    (1810–1882), working in Müller’s laboratory, estab-
    lished that both plants and animals were composed of
    living cells with the nucleus as the essential feature.
    Robert Hooke (1655–1703) earlier had coined the
    word “cell” from the structure he saw in cork. “The
    cells are organisms,” said Schwann, “and animals as
    well as plants are aggregates of these organisms…”
    These two researchers destroyed the existing humoral
    and the competing solidistic concepts of tissue com-
    position. Johannes Müller (1801–1859) was first to
    report that cancers also were composed of living cells.
    In his landmark publication of 1838, Uber den feinen
    Bau und die Formen der krankhaften Geschwülste,
    Müller noted the similarity of cells in a “scirrhus” of
    the breast and its metastases in the ribs and noted that
    cancer cells had lost the proportions of normal cells17

    (Figure 1–4). Rudolph Virchow, also of Berlin,
    Müller’s former student and the founder of cellular
    pathology, is responsible for the dictum that “all cells
    come from cells.” His lectures, Die Cellularpatholo-
    gie, published in 1858, laid to rest the notion of spon-
    taneous generation of living cells from a liquid
    “blastema.” But Virchow did not make the connection
    between migrating malignant cells and metastases; he
    thought that axillary metastases arose from cells in
    the nodes responding to “hurtful ingredients” or “poi-
    sonous matter” from the cancer in the breast.18 Müller
    was perhaps the first to suspect that spread of malig-
    nant cells constituted the mechanism of metastasis,
    later confirmed by the microscopic work of Carl
    Thiersch (1822–1895) and Wilhelm von Waldeyer
    (1836–1921).4 These insights supported the concept
    that breast cancer spread from a local origin.

    Noteworthy clinical observations were also being
    made. Alfred Velpeau was the first to describe breast
    cancer en cuirasse, the deadly form that spreads
    across the chest like a breast plate.19 Velpeau’s Traite
    des maladies du sein, published in 1854, was a com-
    prehensive review of breast disease of the time.
    Across the English Channel in London, Sir James

    Paget made a brief (1,050 words) but enduring report
    in 1874 describing changes on the nipple that pre-
    ceded breast cancer and continue to bear his name.
    He said, “… certain chronic affections of the skin of
    the nipple and areola are very often succeeded by the
    formation of sirrhous cancer in the mammary
    gland…within at the most two years, and usually
    within one year.”20 Paget’s observation remains as
    valuable today as when it was first made.

    Charles Moore (1821–1870) at the Middlesex
    Hospital in London deserves credit for the en bloc
    principle of resection. Moore was convinced that the
    piecemeal mastectomies of his day spread the “ele-
    ments” of cancer in the surgical wound and
    accounted for the local reappearance of cancer in, or
    adjacent to, the scar. In 1867 he published a strong
    argument for removal of the whole breast intact in
    every case.21 He also recommended removal of axil-
    lary nodes and the pectoral muscles if they were
    involved. William M. Banks in Liverpool carried
    mastectomy a step further in 1882 by practicing rou-
    tine removal of axillary lymph nodes.22

    Similar initiatives were occurring in Germany.
    Ernst G.F. Küster (1839–1922) in Berlin was per-
    forming routine axillary clearance and reported that
    it virtually eliminated recurrences in the axilla. In
    1875 Richard von Volkmann (1830–1889) was rou-
    tinely removing the pectoralis major fascia, and
    Küster’s assistant Lothar Heidenhain (1860–1940)
    held the muscle itself suspect. Their microscopic
    studies of mastectomy specimens showed extension

    Figure 1–4. Figure 9, Table I, from Johannes Müller’s Uber den
    feinen Bau und der Formen dei Krankhafte Geschwülste, 1838, illus-
    trating for the first time the cellular structure of breast cancer. Repro-
    duced from Müller J.17

    8 BREAST CANCER

    of cancer to the deep pectoral fascia, and occasion-
    ally to the muscle itself, when it had not been sus-
    pected. Samuel W. Gross at Jefferson Medical Col-
    lege in Philadelphia (1837–1879) attributed a
    3-year survival of 19.4% to routinely removing not
    only the whole breast but the pectoralis fascia and
    axillary contents.4

    The events in Germany influenced William S.
    Halsted (1852–1922), Professor of Surgery at Johns
    Hopkins Hospital in Baltimore, to devise what
    became known as the radical mastectomy. He
    reported the operation in 1894 almost simultane-
    ously with a similar report by Willy Meyer in New
    York.23 Through a large “tear-drop” incision, Hal-
    sted removed en bloc the breast complete with its
    skin, the axillary lymph nodes, “a part, at least” of
    the pectoralis major muscle (the sternal portion) and
    “usually” cleared the supraclavicular region. Halsted
    had adopted this “complete” operation 5 years ear-
    lier, and emphasized that the pectoralis major mus-
    cle must be removed in all cases to obtain a secure
    tumor-free deep surgical margin. He explained that
    von Volkmann had removed the pectoral muscles in
    38 cases with reduction in local recurrences. As
    Moore before him, Halsted wrote that the crux of the
    operation was “to remove in one piece all of the sus-
    pected tissues” lest the wound become infected by
    the division of tissues or lymphatics invaded by the
    disease, and lest shreds or pieces of cancerous tissue
    be overlooked in a piecemeal extirpation. It is clear
    from Halsted’s descriptions that the supraclavicular
    clearance initially was a removal of tissues superior
    to the axillary vessels in the course of the axillary
    dissection, later a formal dissection through a cervi-
    cal incision and ultimately an excursion that he
    abandoned. The complete operation resulted in a
    large open wound left to heal by granulation. Two
    years later he began to close the wound with a split-
    thickness skin graft, a technique developed by
    Thiersch (Figure 1–5). Eventually primary closure
    of skin became more popular.

    Whether radical mastectomy resulted in
    improved local control is unclear. Halsted recog-
    nized “local” as recurrences in the surgical area
    within 3 years of operation (6% in his cases);
    “regionary” recurrences, by his definition, appeared
    after 3 years or in the skin away from the scar. The

    German literature did not distinguish local from
    regionary recurrences. By counting both, and mak-
    ing the unlikely assumptions that cases were compa-
    rable and had equal periods of follow-up, his recur-
    rence rate was 20% compared to 55 to 82% for his
    German counterparts. Halsted disregarded that local
    and/or regionary recurrences totaled 58% when von
    Volkmann had removed the pectoralis major muscle
    and 60% when he had not, a negligible difference.

    Halsted had no surgical deaths despite having
    “old” patients. “Their average age is nearly
    55 years,” he said. “They are no longer very active
    members of society.” This comment is strange to
    modern ears, but the average life span at the time
    was 47 years. For fear of spreading cancer with a
    biopsy, diagnosis was almost always clinical, estab-
    lished histologically after the operation. In doubtful
    cases Halsted said, “The excision of a specimen for

    Figure 1–5. William S. Halsted’s radical mastectomy. A case pic-
    tured in 1912. The operation resulted in a large wound closed with
    skin grafts. Reproduced with permission from the Classics of
    surgery library. Surgical papers. William Stewart Halsted. Vol. 2.
    Special Edition. Birmingham (AB): L. B. Adams Jr. 1984; Figure 2,
    Plate LX. p. 82.

    History of Breast Cancer 9

    macroscopic or microscopic is never resorted to
    except just before operation.”24

    Radical surgery had intellectual support from
    W.S. Handley’s theory of permeation, which held
    that breast cancer spread centrifugally in continuity,
    and lymph nodes provided mechanical barriers.
    Blood vascular spread was insignificant; tumor
    emboli were destroyed by clot.25 Halsted’s operation
    was used, with mixed results, for the next 80 years.

    Cushman D. Haagensen (d 1990) was both a
    staunch supporter and a critic of radical mastectomy.19

    His book Diseases of the Breast, published in 1956, is
    a classic. Haagensen’s careful analysis of cases treated
    at Presbyterian Hospital in New York City resulted in
    eight “criteria of inoperability” to discourage inappro-
    priate use of the operation. He also standardized phys-
    ical breast examination and originated the Columbia
    Clinical Classification (CCC) staging system. After
    the CCC, staging systems became increasingly sophis-
    ticated and eventuated in the current Tumor, Node,
    Metastasis system initially adopted in 1954 by the
    International Union Against Cancer. Prior to random-
    ized clinical trials, staging provided the principal
    means for comparing different methods of treatment.

    As the nineteenth century came to a close, mas-
    tectomy appeared better than no treatment but still
    cured less often than not. The actuarial survival of
    Halsted’s first fifty cases 5 years from the first
    symptom (40.4%) was greater than twice that of
    untreated patients in the Middlesex Hospital Charity
    Ward in London admitted between 1805 and 1933,
    which was 18%.26,27

    Two events of this time were momentous for the
    future treatment of breast cancer. The first was the
    discovery of x-rays, and the second was the discov-
    ery that breast cancer was hormone dependent. Dis-
    covery of x-rays by Wilhelm Conrad Röntgen in
    Würzburg in 1895 provided the basis for radiother-
    apy and mammography. The mysterious ray, desig-
    nated “x,” not only penetrated tissues but also killed
    cancers. One year after Röntgen’s discovery, x-rays
    were used to treat three cases of breast cancer, two
    by Hermann Goeht in Hamburg and one by Emile
    Herman Grubbé in Chicago.4 All three had
    advanced, inoperable cancers and died shortly after-
    wards. With the development of dosimetry, improve-
    ments in instrumentation and appropriate safe-

    guards, radiation therapy became an effective local
    treatment of inoperable cancers, and a postoperative
    (and sometimes preoperative) supplement to mas-
    tectomy that ultimately enabled breast-conserving
    surgery. The discovery of radium in 1898 by Pierre
    and Marie Curie added interstitial radiation to thera-
    peutic options. Geoffrey Langdon Keynes in Lon-
    don (1932) used radium as the sole treatment of
    operable cases.28 The obvious benefit of ionizing
    irradiation was in reducing the bulk of large cancers
    and in reducing recurrence in treated fields. An
    inferred influence on survival proved elusive.

    The hormonal treatment of breast cancer began
    with oophorectomy. In 1899 Albert Schinzinger
    (1827–1911) in Freiburg commented on the poor
    prognosis of young women with breast cancer and
    proposed castration to age them and slow down the
    malignant growth. Independent of this suggestion,
    George Thomas Beatson (1848–1933) of Glasgow
    performed the first castration for breast cancer
    7 years later. Beatson knew by his studies of lactation
    that castration or rebreeding of cows shortly after
    they calved prolonged milk production, both mea-
    sures having in common the interruption of ovarian
    function. Since the hyperplastic cells of lactation
    decomposed into milk, he reasoned that castration
    might make the hyperplastic cells of breast malig-
    nancy do so as well. The reasoning was wrong, but
    the result was gratifying. In 1896 he reported tempo-
    rary tumor regression after oophorectomy in three
    cases of advanced breast cancer. Beatson’s discovery
    established the palliative value of oophorectomy, and
    for a period it became a regular adjuvant to mastec-
    tomy by some surgeons.29 Secondary endocrine
    surgery with adrenalectomy and hypophysectomy
    developed as sequels to oophorectomy, but in time
    endocrine surgery was replaced by hormone therapy
    (Henry Starling described hormones in 1905) and,
    ultimately, by pharmacologic methods of reducing
    estrogen production or its effects with luteinizing
    hormone-releasing hormone agonists, estrogen
    receptor modulators and aromatase inhibitors. Dis-
    covery of intracellular estrogen receptors (ER) in
    breast cancers by Elwood Jensen in Chicago in 1967
    was another milestone in hormone therapy, permit-
    ting patients who could benefit from hormone ther-
    apy to be distinguished from those who could not.30

    10 BREAST CANCER

    TWENTIETH CENTURY

    The next 100 years resulted in a retreat from radical
    surgery and the introduction of mammography and
    chemotherapy. Research confirmed a hereditary
    component of breast cancer. As important as all else
    came a demand for scientific evidence to support
    claims of efficacy and to supplant the anecdotes and
    polemics of the past. Cooperative groups of clini-
    cian investigators amassed large numbers of patients
    for study, and randomized, controlled clinical trials
    with sophisticated statistical analysis of data became
    commonplace. Breast cancer was recognized as a
    major health problem in the Western world, stimu-
    lating a concerted effort against it.

    In the early decades, many sought to improve the
    results of radical surgery with “extended” radical
    mastectomies. Margottini and Veronesi in Milan,
    Caseres in Peru, and Urban and Sugarbaker in the
    United States removed the internal mammary nodes.
    Dahl-Iverson in Copenhagen removed the supra-
    clavicular and internal mammary nodes and Wan-
    genstein in Minnesota added removal of mediastinal
    nodes. Other than showing that extra-axillary nodes
    often contained metastases and that their removal
    improved regional tumor control, cures were not
    increased, and these extensions were eventually
    abandoned in favor of chest wall and regional irradi-
    ation. As Handley’s permeation theory lost cre-
    dence, D. H. Patey and R. S. Handley in London felt
    justified in preserving the pectoralis major muscle
    unless it was directly involved by cancer, an opera-
    tion they called the “conservative” radical mastec-
    tomy. With the support of surgeons in the United
    States such as Hugh Auchincloss Jr. in New York,
    this operation eventually prevailed in 1979 as the
    “modified” radical mastectomy.31

    Mammography, unarguably the most important
    advance to date in the detection of breast cancer,
    developed in parallel with surgery. Even early physi-
    cians had recognized that small breast cancers were
    the most curable. Mammography allowed many
    breast cancers to be detected when clinically occult,
    including ductal carcinoma in situ, which was regu-
    larly curable. Film-screen mammography involved
    penetrating the breast with x-rays to activate a rare
    earth screen that glowed in response. This screen

    exposed a transparent, photosensitive film in the
    same cassette which, when developed, provided an
    image in various shades of gray for interpretation. In
    Robert Egan’s History of Mammography, he gives
    Stafford L. Warren at Rochester Memorial Hospital
    in Rochester, New York, credit for early explorations
    of mammography beginning in 1926 but also men-
    tioned that the German surgeon, Albert Salomon,
    performed studies with radiographs of breasts
    resected for carcinoma as early as 1913 before his
    work was apparently interrupted by World War I.32

    The technique met resistance despite such advocates
    as Jacob Gershon-Cohen in Philadelphia and
    Charles M. Gros in Strasbourg until Egan, while a
    radiologist at M. D. Anderson Hospital in Houston,
    Texas, developed the soft tissue technique that
    allowed mammography to move forward.

    An early randomized trial of screening with
    mammography and physical examination in New
    York by Sam Shapiro and Philip Strax in 1963
    demonstrated that 30% of cancers could be detected
    by mammography alone, and deaths from cancers
    among screened women were reduced 30% com-
    pared with unscreened. After a host of radiologists
    was trained in the technique of mammography, a
    demonstration project, the Breast Cancer Detection
    Demonstration Project (BCDDP), begun in 1973
    and sponsored by the National Cancer Institute and
    the American Cancer Society (ACS), screened
    283,222 asymptomatic women. The BCDDP estab-
    lished the feasibility of mass population screening.
    Multiple randomized clinical trials of screening fol-
    lowed, showing that regular mammograms could
    detect 85 to 90% of asymptomatic breast cancers
    with a reduction of breast cancer mortality. Periodic
    mammograms and physical examinations for detec-
    tion of breast cancer in asymptomatic women 40
    years of age and older received endorsement by the
    NCI, ACS, and numerous professional groups.33

    Mammography was followed by a number of
    innovative means for imaging the breast. Xeromam-
    mography appeared briefly.34 This dry-process tech-
    nique recorded all structures in the breast with
    equally good detail and could be examined without
    view boxes, but it disappeared from use after further
    improvements in film-screen mammography. Endur-
    ing adjuncts to mammography were ultrasonography

    History of Breast Cancer 11

    and magnetic resonance imaging (MRI). Ultrasonog-
    raphy came into use in the 1950s. As well as allow-
    ing for the distinction between cysts and solid
    masses, it could characterize solid masses and per-
    mitted irradiation-free, real-time, guided needle
    biopsy of suspicious lesions. Malignant lesions
    detected by other means were not always visible on
    ultrasonography, and results were highly operator-
    dependent, making it unsuited for population screen-
    ing. MRI proved valuable in special situations.

    As the twentieth century advanced, opposition to
    radical surgery grew. Kaae and Johansen in Denmark
    and Robert McWhirter in Scotland maintained that
    simple mastectomy with regional irradiation was the
    equal of radical mastectomy, and preferable.35

    McWhirter protested that the selective use of radical
    mastectomy made the results look better but offered
    no overall increase in cures. George Crile Jr. in
    Cleveland argued for conservative surgical treatment
    based on a biological view of breast cancer, largely
    immunologic.36 Most compelling, however, was that
    radical surgical removal of tissues had reached its
    limits with no decrease in mortality rates. In 1939
    Gray showed that early lymphatic spread to axillary
    nodes was by embolism rather than by permeation,
    and blood vascular spread was increasingly accepted
    as the mechanism of general dissemination.37,38

    Bernard Fisher, Professor of Surgery at the Uni-
    versity of Pittsburgh and a researcher in the biology of

    metastasis, became the intellectual leader and the
    most compelling spokesman for the need to critically
    re-evaluate the treatment of breast cancer. Fisher’s
    laboratory investigations indicated that lymph nodes
    were not effective barriers to cancer spread. Referring
    to Halsted’s rationale for radical mastectomy, Fisher
    wrote in 1970 that, “…either the original surgical
    principles have become anachronistic or, if they are
    still valid, they were conceived originally for the
    wrong reasons.”39 Much like a modern Galen, Fisher
    asserted that breast cancer was a systemic disease and
    that its course was determined by a biologic struggle
    between tumor and host. Fisher implied that viable
    cancer cells always, or almost always, disseminated
    before diagnosis. His thesis presented two testable
    hypotheses: (1) variations in local treatment were
    unlikely to influence cure, and (2) effective systemic
    treatment was necessary to improve cure rates. As
    Chairman of the National Surgical Adjuvant Breast
    and Bowel Project (NSABP), Fisher was able to
    implement large, randomized, controlled clinical tri-
    als to test these concepts and to stimulate others to do
    the same (Figure 1–6). The results confirmed the
    observations of Moore, Küster, and Halsted, namely,
    that limited operations resulted in poor local and
    regional control, and that patients with recurrence
    fared poorly.40–42 As predicted, they also confirmed
    that whether the regional nodes or the whole breast
    were removed, overall cure rates among different

    Figure 1–6. Bernard Fisher MD, modern researcher in the biology of breast cancer who revised Halstedian concepts, (fourth from the right
    in the front row) with early members of the NSABP at a group meeting in Florida, May 1978.

    12 BREAST CANCER

    treatment groups proved similar. The explanation
    offered was that failure of local control indicated
    incurability at the outset. But the need to retreat (“sal-
    vage”) was distressing for all, and as local or regional
    recurrence might jeopardize cure for some, optimum
    tumor control at the outset remained a priority.

    The greatest impact of these trials was on man-
    agement of the breast itself. As confidence grew in
    irradiation for controlling occult regional metas-
    tases, the question was whether irradiation could do
    the same for occult tumor in the breast. Selected
    cases so treated by F. Baclesse in France, Ruth
    Guttman in the United States, Sakan Mustakallio in
    Finland, and others had suggested this was the case
    as early as 1965.43 After an initial but unsatisfactory
    beginning at Guy’s Hospital in London, controlled
    trials of breast conservation started in Milan, Italy,
    in 1973 by Umberto Veronesi and by the NSABP in
    1976.44,45 These trials established that excision of the
    primary tumor, “lumpectomy,” followed by whole
    breast irradiation was as effective as total mastec-
    tomy for both local and ultimate disease control of
    most early-stage cases and was an obvious cosmetic
    improvement. Based on these outcomes, in 1990 the
    NCI sanctioned breast-conserving surgery as the
    preferred treatment of stage I and II breast cancers.46

    Axillary sentinel lymph node biopsy (SLNB)
    was rapidly adopted after it was introduced in 1997,
    making routine axillary lymph node dissection
    unnecessary. Axillary dissection could be reserved
    instead for cases in which the SLNB showed nodal
    metastases, thereby sparing many the morbidity of
    this operation.47 Surgical treatment of the breast and
    the regional nodes could be customized to individual
    needs, and with the combination of SLNB and
    breast conservation, the surgical component of mul-
    tidisciplinary treatment reached a minimum.

    Chemotherapy developed in parallel with changes
    in local treatment. Its beginnings can be traced to the
    use of mustard gas in World War I. Exposure caused
    depression of bone marrow and lymphoid tissue fol-
    lowed by death from pneumonia. The effects on tis-
    sues were similar to those of ionizing radiation and
    suggested usefulness against lymphomas. Experi-
    ments with animals followed, and, indeed, nitrogen
    mustard produced regression of implanted lymphoma
    in mice. In 1942 it was first used to treat human lym-
    phoma at Yale University; the results of which were

    not reported by Goodman and Philips until 1946, a
    delay necessitated by the need for wartime secrecy.
    Reference is sometimes made in texts to events sur-
    rounding explosion of mustard agent (dichloroethyl
    sulfide) bombs aboard the S.S. John Harvey on
    December 2, 1943, in Bari Harbor, Italy, during WW
    II as the stimulus for research into chemotherapy, but
    this event followed the clinical investigations at Yale
    University.48 Continued development produced such
    therapeutically useful alkylating agents as busulfan,
    cyclophosphamide and chlorambucil. Additional
    agents with various mechanisms of cytotoxicity fol-
    lowed. None proved toxic specifically for cancer cells
    or free of undesirable side effects, and none cured
    overt breast cancers, but their judicious use proved
    clinically useful. Systemic “chemotherapy,” a word
    coined by the researcher Paul Erlich, often produced
    temporary regression and occasionally complete dis-
    appearance of advanced breast cancers.49 Initial trials
    of intravenous, perioperative triethlylene-thiophos-
    phoramide (Thio-TEPA) in the late 1950s, intended to
    destroy tumor cells released during mastectomy, were
    failures, but extended adjuvant treatment with
    L-phenylalanine mustard directed against occult
    micrometastases improved the survival of patients
    with early stage breast cancer.50,51 A similar approach
    using combinations of drugs with different mecha-
    nisms of action (eg, cyclophosphamide, fluorouracil,
    and methotrexate (CMF), and doxorubicin combina-
    tions) proved more effective, securing adjuvant
    chemotherapy an established place in multidiscipli-
    nary treatment. With the addition of chemotherapy,
    treatment of breast cancer truly became a coordi-
    nated effort of specialists, bringing to bear a medley
    of surgery, radiation therapy, and systemic chemo-
    hormonal therapy on the local and systemic compo-
    nents of the disease.

    As the twentieth century closed, breast cancer was
    recognized as a disorder of unrestrained cell growth,
    but its instigation remained an enigma. A virus caused
    the disease in mice, but apparently not in humans;
    ingestion of aromatic hydrocarbons (dimethylbenzan-
    thracene) produced it in rats. In humans, exposure to
    ionizing radiation increased risk, as evidenced in sur-
    vivors of the atomic bombing of Hiroshima during
    WW II and the recipients of multiple fluoroscopies
    incident to treatment of pulmonary tuberculosis, infor-
    mation spurring closer regulation of mammography

    History of Breast Cancer 13

    and other radiological procedures. Hormone replace-
    ment therapy to alleviate menopausal symptoms also
    increased risk, prompting cautions about exposure to
    exogenous estrogens.52 The discovery of predisposing
    mutations in BRCA1 and BRCA2 genes of families
    prone to breast cancer confirmed genetic transmis-
    sion and provided a means to identify individuals at
    great risk.53,54 Among preventive strategies, early cas-
    tration was effective but unacceptable; in 1998
    tamoxifen, a synthetic estrogen receptor modulator,
    became the first drug proven to lower risk and the
    first approved for this use.55 Prophylactic mastec-
    tomies offered almost total protection, and became an
    option for women especially in need.56

    Breast cancer remained a daunting problem as
    science and medicine reached the third millennium
    AD, but a problem more accurately defined than ever
    before and upon which all the tools of modern science
    were brought to bear. Research explored cellular
    growth factors and intracellular signaling pathways
    that might be exploited against it. For practicing
    physicians radiotherapy, medical oncology, surgical
    oncology, and even breast surgery had become spe-
    cialties. Cancer institutes dotted the country. For the
    record, in the United States in 2004 an estimated
    217,000 women continued to develop breast cancer
    each year and 40,000 died of it annually. With screen-
    ing and modern therapy, the death rate had begun to
    decline and overall relative survival 5 years after diag-
    nosis, cured and uncured, was 86.6%.57

    COMMENT

    The sometimes heroic, often tragic, and always
    poignant story of breast cancer is incomplete; happy
    will be the day when the final chapter is written.
    When that day comes, it may not get the attention it
    deserves. It will come in familiar voices on the
    nightly news: “Today doctors at (some) medical cen-
    ter announced that a (vaccine?) prepared from the
    (prions?) of breast cancer resulted in immediate and
    total disappearance of all signs of the disease in
    eleven advanced cases. Further studies are planned
    to follow up this promising development. In interna-
    tional news…” Reactions will be mixed. Most will
    notice without comment. Skeptics will quip, “Yeah,
    another breakthrough!” But it will be true. Others,
    robbed of loved ones, will hesitate in melancholy

    reflection. More than suspected will reap the
    rewards, and after more than 5,000 years of telling,
    the story of breast cancer will have been told.

    ACKNOWLEDGMENT

    The author wishes to thank Judith H. Donegan, MD,
    PhD, for constructive criticism of the manuscript.

    REFERENCES

    1. Encyclopedia Britannica, 15th ed. Encyclopedia Britannica,
    Inc. Chicago: Encyclopedia Britannica Inc.; 1978.
    Macropedia. Vol 11 p. 823.

    2. Breasted JH, editor. The Edwin Smith Surgical Papyrus.
    Chicago, IL: The University of Chicago Press; 1930, Special
    Edition. 1984. The Classics of Surgery Library. Division of
    Gryphon Editions, Ltd. Birmingham (AB). Frontispiece.

    3. Homer. Iliad. Translated by WHD Rouse. New York: A Signet
    Classic. New American Library; 1966. p. 36.

    4. De Moulin D. A short history of breast cancer. Boston: Mar-
    tinus Nijhoff; 1983. p. 1–107.

    5. Lewison EF. Saint Agatha the patron saint of diseases of the
    breast in legend and art. Bull History of Medicine
    1950;24:409–20.

    6. Lyons AS, Petrucelli RJ. Medicine. An illustrated history.
    New York: Harry N. Abrams Publishers; 1978. p. 294–317.

    7. Hoxey HM. You don’t have to die. New York: Milestone
    Books Inc; 1956. p. 47.

    8. Yalom M. A history of the breast. New York: Alfred A.
    Knopf; 1997. p. 211.

    9. LeDran HF. Memoires avec un précis de plusieurs observa-
    tions sur le cancer. Memories de l’academie royale de
    chirurgie 1757;3:1–54.

    10. Saunders JB deC M, O’Malley CD. The anatomical drawings
    of Andreas Vesalius. New York: Bonanza Books; 1982.
    p. 172–3.

    11. Cooper AP. The anatomy and diseases of the breast. Philadel-
    phia: Lea and Blanchard; 1845.

    12. Sappey MPC. Anatomie, physiologie, pathologie des vais-
    seaux lymphatique considérés chez l’homme et les
    vertébrés. Paris: A Delahaye and E Lecrosnier; 1874.

    13. Lillberg K, Verkasalo PK, Kaprio J, et al. Personality charac-
    teristics and the risk of breast cancer: a prospective cohort
    study. Int J Cancer 2002;100 361–6.

    14. Robbins G, editor. Silvergirl’s surgery—the breast. Austin:
    Silvergirl Inc.; 1984. p. 25–9.

    15. Garrison FH. An introduction to the history of medicine. 4th
    ed. Philadelphia: WB Saunders, Co.; 1929. p. 588–9.

    16. Encyclopedia Britannica, 15th ed. Chicago: Encyclopedia
    Britannica Inc.; 1978. Macropedia Vol 11 p. 837.

    17. Müller J. Uber den feinen Bau und der Formen dei
    Krankhafte Geschwülste. Berlin: G Reimer; 1838.

    18. Virchow R. Cellular pathology. Birmingham; The Classics of
    Medicine Library, Division of Gryphon Editions, Ltd.;
    1978. p. 66.

    19. Haagensen CD. Diseases of the breast, 2nd ed. Philadelphia:
    W. B. Saunders Co; 1971. p. 394–5.

    14 BREAST CANCER

    20. Paget J. On disease of the mammary areola preceding cancer
    of the mammary gland. St. Bartholomew Hospital
    Reports;1874:vol. 10; p. 75–8.

    21. Moore C. On the influence of inadequate operations on the
    theory of cancer. Royal Medical and Chirugical Society.
    London. Med Chir Trans 1867;32:245–80.

    22. Banks WM. Free removal of mammary cancer with extirpa-
    tion of the axillary glands as a necessary accompaniment.
    Paper read before the British Medical Association at
    Worcester. 1882.

    23. Halsted WS. The results of operations for the cure of cancer of
    the breast performed at the Johns Hopkins Hospital from
    June 1889 to January 1894. Johns Hopkins Hospital
    Reports. Baltimore 1894–95;4:297–350.

    24. Halsted WS. The results of radical operations for the cure of
    cancer of the breast. Trans Am Surg Assoc 1907;25:61–79.
    Reprinted in: Surgical papers of William Stuart Halsted
    Birmingham (AL): Gryphon Editions; 1984. p 80.

    25. Handley WS. Cancer of the breast and its operative treat-
    ment. London: John Murray; 1906.

    26. Bloom, HJG, Richardson WW, Harries EJ. Natural history of
    untreated breast cancer (1805-1933). Comparison of
    untreated and treated cases according to histological
    grade of malignancy. Brit Med J 1962;I:213–21.

    27. Donegan WL. Staging and prognosis. In: Donegan WL,
    Spratt JS, editors. Cancer of the breast, 5th ed. Philadel-
    phia: W.B. Saunders Co; 2002. p. 478.

    28. Keynes GL. The radium treatment of carcinoma of the breast.
    Brit J Surg 1942;19:415–80

    29. Horsley JS III, Horsley GW. Twenty years experience with
    prophylactic bilateral oophorectomy in the treatment of
    carcinoma of the breast. Ann Surg 1962;155:935.

    30. Jensen EV, DeSombre ER, Jungblut PW. Estrogen receptors
    in hormone responsive tissues and tumors. In: Wissler
    RW, Dao TL, Wood S Jr., editors. Endogenous factors
    influencing host-tumor balance. Chicago: University of
    Chicago Press; 1967.

    31. Special Report: Treatment of primary breast cancer. N Engl
    J Med 1979;301:340.

    32. Egan RL. Mammography, 2nd ed. Springfield: Charles C
    Thomas; 1972. p. 3–22.

    33. NCI statement on mammography screening. Available at: http://
    www.cancer.gov/newscenter/mammstatement31jan02
    (accessed Jan 4, 2005).

    34. Wolfe JN. Xeroradiography of the breast. Springfield:
    Charles C Thomas; 1972. p. 3–5.

    35. McWhirter R. Simple mastectomy and radiotherapy in the
    treatment of breast cancer. Br J Radiol 1955;28:128.

    36. Crile G Jr. A biological consideration of treatment of breast
    cancer. Springfield, IL: Charles C. Thomas;1967.

    37. Gray JH. The relation of lymphatic vessels to the spread of
    cancer. Br J Surg 1939;26:462.

    38. Ewing J. Neoplastic diseases, 4th ed. Philadelphia: W. B.
    Saunders Co; 1940. p. 63–74.

    39. Fisher B. The surgical dilemma in the primary therapy of
    invasive breast cancer: a critical appraisal. Current prob-
    lems in surgery. Chicago: Year Book Medical Publishers
    Inc.;1970.

    40. Fisher B, Montague E, Redmond C, et al. Comparison of rad-

    ical mastectomy with alternative treatments for primary
    breast cancer. Cancer 1977;39:2827–39.

    41. Veronesi U, Valagussa P. Inefficacy of internal mammary node
    dissection in breast cancer surgery. Cancer 1981;47:170–5.

    42. Fisher B, Redmond D, Poisson R, et al. Eight-year results of
    a randomized clinical trial comparing total mastectomy
    and lumpectomy with or without irradiation in the treat-
    ment of breast cancer. N Engl J Med 1989;320:822–8.

    43. Baclesse F. Five-year results in 431 breast cancers treated
    solely by roentgen rays. Ann Surg 1965;61:103–4.

    44. Veronesi U, Volterrani F, Luini A, et al. Quadrantectomy ver-
    sus lumpectomy for small size breast cancer. Eur J Can-
    cer 1990;26:671–3.

    45. Fisher B, Anderson S, Redmond CK, et al. Reanalysis and
    results after 12 years of follow-up in a randomized clinical
    trial comparing total mastectomy with lumpectomy with
    or without irradiation in the treatment of breast cancer. N
    Engl J Med. 1995;333:1456–61.

    46. Treatment of Early-Stage Breast Cancer. NIH Consens State-
    ment Online 1990 Jun 18–21 [cited 2005 October 26];8:
    1–19.

    47. Guilliano AE, Jones RC, Brennan M, Statman R. Sentinel
    lymphadenectomy in breast cancer. J Clin Oncol 1997;
    15:2245–50.

    48. Papac RJ. Origins of cancer therapy. Yale J Biol and Med
    2002;74:391–8.

    49. DeVita VT. Principles of chemotherapy. In: DeVita VT Jr,
    Hellman S, Rosenberg SA, editors. Cancer—principles
    and practice of oncology. Philadelphia: J. B. Lippincott
    Co; 1982. p. 132–3.

    50. Noer RJ. Adjuvant chemotherapy. Thio-tepa with radical
    mastectomy in the treatment of breast cancer. Am J Surg
    1963;106:405–12.

    51. Fisher B, Fisher ER, Redmond C. Ten-year results from the
    National Surgical Adjuvant Breast and Bowel Project
    (NSABP) clinical trial evaluating the use of L-phenylala-
    nine mustard (L-PAM) in the management of primary
    breast cancer. J Clin Oncol 1986;4:929–41.

    52. Chlebowski RY, Hendrix SL, Langer RD, et al. Influence of
    estrogen plus progestin on breast cancer and mammography
    in healthy postmenopausal women: the Women’s Health Ini-
    tiative Randomized Trial. JAMA 2003;289:3243–53.

    53. Friedman LS, Ostermeyer EA, Szabo CI, et al. Confirmation
    of BRCA1 by analysis of germline mutations linked to
    breast and ovarian cancer in ten families. Nat Genet
    1994;8:399–404.

    54. Futreal PA, Liu Q, Shattuck-Eidens D, et al. BRCA1 muta-
    tions in primary breast and ovarian carcinomas. Science
    1994;266:120–2.

    55. Fisher B, Constantino JP, Wickerham DL, et al. Tamoxifen
    for prevention of breast cancer: report of the National
    Surgical Adjuvant Breast and Bowel Project P-1 Study.
    J Natl Cancer Inst 1998;90:1371–88.

    56. Hartmann LC, Schaid DJ, Woods JE, et al. Efficacy of bilat-
    eral prophylactic mastectomy in women with a family his-
    tory of breast cancer. N Engl J Med 1999;340:77–84.

    57. American Cancer Society 2004 statistics. Available at:
    http://www.cancer.org/downloads/MED/Page4
    (accessed Jan 11, 2005).

      BREAST CANCER

    • Copyright
    • Dedication
    • Contents
    • Preface
    • Contributors
    • Ch01: History of Breast Cancer
    • Ch02: Anatomy of the Breast, Axilla, and Thoracic Wall
    • Ch03: The Evolving Concept of the Breast Cancer
    • Ch04: Epidemiology of Breast Cancer
    • Ch05: Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer
    • Ch06: Molecular Basis of Breast Cancer
    • Ch07: Breast Cancer Risk Assessment and Management
    • Ch08: Role of Screening in Breast Cancer Mortality Reduction
    • Ch09: Diagnostic Breast Imaging
    • Ch10: Sonography of Breast Cancer
    • Ch11: Magnetic Resonance Imaging
    • Ch12: Diagnostic Techniques
    • Ch13: Pathology of Invasive Breast Cancer
    • Ch14: Staging and Histologic Grading
    • Ch15: Ductal Carcinoma In Situ
    • Ch16: Oncoplastic Surgery of the Breast
    • Ch17: Evaluation and Surgical Management of Stage I and II Breast Cancer
    • Ch18: Locally Advanced Breast Cancer
    • Ch19: Axillary Staging and Therapeutics
    • Ch20: Adjuvant Chemotherapy
    • Ch21: Endocrine Therapy of Early and Advanced Breast Cancer
    • Ch22: Radiation Therapy in Early and Advanced Breast Cancer
    • Ch23: Evolution in Breast Reconstruction
    • Ch24: Unusual Breast Histology
    • Ch25: Multifocal, Multicentric, and Bilateral Breast Cancer
    • Ch26: Breast Cancer in the Previously Augmented Breast
    • Ch27: Breast Cancer in the Irradiated Breast
    • Ch28: Novel Radiation Therapy Techniques
    • Ch29: Novel Radiation Therapy Techniques
    • Ch30: Hormone Therapy and Breast Cancer
    • Ch31: Male Breast Cancer
    • Ch32: Management of Locoregional Recurrences
    • Ch33: Breast Cancer and Multiethnic/Multiracial Populations
    • Ch34: Image-Guided Ablation for Breast Cancer
    • Ch35: Lymphedema
    • Ch36: Surveillance Strategies for Breast Cancer Survivors
    • Ch37: A Patient’s Perspective
    • Index
    • Exit

    1

    R
    ev
    ie
    w

    Published: 08/06/2017 Received: 20/03/2017

    ecancer 2017, 11:746 https://doi.org/10.3332/ecancer.2017.746

    Copyright: © the authors; licensee ecancermedicalscience. This is an Open Access article distributed under the terms of the Creative
    Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction
    in any medium, provided the original work is properly cited.

    The past and future of breast cancer treatment—from the papyrus to
    individualised treatment approaches
    Felipe Ades1, Konstantinos Tryfonidis2 and Dimitrios Zardavas3

    1Hospital Albert Einstein, Avenida Albert Einstein, 627 – Morumbi, São Paulo – SP, 05652-900 Brazil
    2European Organisation for Research and Treatment of Cancer, Avenue E. Mounier 83/11, 1200 Brussels, Belgium
    3Breast International Group (BIG), Boulevard de Waterloo 76, Brussels 1000, Belgium

    Correspondence to: Felipe Ades. Email: felipeades@gmail.com and felipe.ades@einstein.br

    Abstract

    Cancer is one of the oldest diseases ever described, since ancient Egypt there have always been attempts to treat and cure this illness.
    The growing body of knowledge about breast cancer biology and improvements in surgical and medical treatments has been built over time
    with contributions from many talented and enthusiastic physicians and researchers. Medical advances have changed the approach from a
    previously incurable condition, into a surgical disease. Further improvements in cancer biology have allowed the development of systemic
    treatments, hormonal therapies, and targeted drugs. The description of the molecular intrinsic subtypes of breast cancer clarified the under-
    standing of breast cancer as a group of heterogeneous diseases, associated with different clinical outcomes, and therapeutic opportunities.
    This paper reviews how breast cancer treatment has improved since the earliest descriptions, in ancient times, and how future approaches,
    such as gene signatures, molecular profiling, and liquid biopsies, aim to further develop individualised treatments and improve treatment
    outcomes.

    Keywords: breast cancer, cancer history, breast surgery, gene signature, molecular profiling, liquid biopsy

    R
    ev
    ie
    w

    2 www.ecancer.org

    ecancer 2017, 11:746

    Breast cancer treatment in ancient times

    Cancer is one of the oldest diseases ever described by medicine. One of the earliest manuscripts reporting on cancer treatment was the
    Egyptian papyrus of Edwin Smith, dated 1600BC but possibly a copy from a much older document, from 2500 to 3000BC [1]. It is consid-
    ered to be the first known medical treatise, as it embraces a rational and scientific approach towards medical treatments, in contrast with
    older manuscripts that reported magic and other mystical methods to treat diseases. The papyrus describes 48 generic clinical cases where
    rudimentary surgical procedures were used to treat wounds, fractures, and cancer, according to the different parts of the body. The part
    dedicated to the breast depicts techniques to treat breast cancer and wounds, but not with the objective to eradicate with curative intent,
    as cancer, at that time, was considered an incurable illness [2].

    Around the year 400BC, the Greek physician Hyppocrates, considered the ‘father of medicine’, created the term cancer. The name derived
    from the word ‘karkinos’, which is the Greek term for crab or crayfish, in an analogy for the invasive behaviour of the disease, touching and
    invading nearby tissues [3]. Others believe that the analogy was due to the similarities between the vascularisation of the tumour and the
    crayfish’s legs. In the second century, the also Greek doctor Galen created the term oncos (the Greek work for swelling) to refer to malig-
    nant diseases. Galen believed that diseases were caused by an imbalance of humours and breast cancer had a systemic nature due to the
    accumulation of black bile in the blood [4].

    From the middle ages to the present day

    With the ascension of the monotheistic religions, little progress in medicine was made in Europe. Early Christian views attributed the cause
    of diseases to God, and treatments, likewise, were based on faith and miracles rather than on surgery and medications. Only in the 10th
    century, with the rise of the Islamic empire, was Greek medicine revived and expanded due to the work of physicians like Ibn Sina [5] and
    Abu Al-Qasim Al-Zahrawi (aka Albucasis) [6]. Accurate translations of the medical manuscripts and the establishment of the early medical
    schools allowed the continuation of medical development [7]. However, the dissection and representation of the human body remained
    forbidden by religious traditions, limiting additional expansion of medical knowledge [8].

    Albucasis, along with the French surgeons Henri de Mondeville [9] and Guy de Chayliac [10], added unique instruments to the surgical
    procedures to remove breast tumours. But it was not until the 16th century that breast surgery flourished. The improvement of surgical
    and medical sciences occurred in the context of the age of enlightenment. Detailed anatomical descriptions made by Adreas Versalius,
    Leonardo da Vinci and others paved the road to a better understanding of the human body allowing the advance of surgical techniques
    [8]. In Scotland, the surgeon John Hunter [11] established the initial concept of staging; ‘if the tumour is moveable, there is no impropriety
    in removing it’ [12]. Nevertheless, surgical approaches were limited due to technology issues; until the mid-19th century, anaesthesia was
    not yet developed for surgical procedures [13]. Surgeons had to rely on technique and, mostly, speed to perform tumour resections. In
    parallel, investigations on a wide range of carcinogens suspected to be involved in breast cancer development and progression started to
    be conducted [7]. For the first time, medicine aimed to cure breast cancer.

    19th century—the beginning of modern breast cancer treatment

    The 19th century saw great improvements in the understanding of the disease and its mechanisms. Observations made by Virchow [14]
    and others underpinned the cellular nature of cancer cells and its differences from their healthy counterparts. This observation dismissed
    many of the previous humoural theories as causes of cancer and its metastasis. Metastatic spread was due to the dissemination of these
    cells by the lymphatic and blood vessels.

    A range of new surgical approaches were developed given the growing body of information on carcinogenesis and metastasis mechanisms
    and important improvements were made on surgical techniques, such as anaesthesia, surgical gloves and vestments, and disinfection.

    In particular, anaesthesia [13] and disinfection allowed, for the first time, surgical freedom to perform wider resections. The en bloc resec-
    tion was experimented by a range of skilled surgeons such as Charles Moore [15], in the United Kingdom, and Kuster and Volkmann [16],

    R
    ev
    ie
    w

    3 www.ecancer.org

    ecancer 2017, 11:746

    in Germany. Axillary dissection was used as part of the surgical treatment of breast cancer by Banks in Liverpool [17]. In 1894, the surgeon
    William Halsted [18] described the surgery that became the standard of care for many years to come, the radical Halsted mastectomy.
    Believing that the disease had an initial local behaviour with a constant path of dissemination, from the breast to the axillary nodes, and
    only then to distant locations of the body, Halsted developed a technique that would remove the whole tumour in one piece along with the
    pectoral muscles, lymphatic vessels, and the axillary lymph nodes [18]. The one piece removal would avoid unattended cancerous tissues
    being left behind and no lymphatic vessels would be damaged, thus no contamination of healthy tissue by cancer cells would occur [19].
    Despite the radical approach and the profound side effects of this wide procedure, this was the first time in history that breast cancer could
    be systematically and scientifically cured.

    Modern times—more improvements and fewer morbidities

    In the 20th century, a rapid expansion in medical knowledge was made, with advances seen in multiple areas of medicine. As with surgery,
    improvements in radiotherapy, chemotherapy, and endocrine therapy were preceded by an accumulation of knowledge about the patho-
    logic mechanisms of the disease.

    The surgeon Thomas Beatson, experimenting with animal models, observed that breast tumours in these animals regressed after oopho-
    rectomy [20]. The relation of sexual hormones with breast cancer was only elucidated later, in 1967, when Elwood Jensen described the
    oestrogen receptor, paving the road for the development of a range of oestrogen-modulating drugs [21].

    The development of chemotherapy also happened in the second half of the 20th century; first with the mustard gas derivatives [22], followed
    by a rapid expansion in the chemotherapy portfolio after the heavy financing of cancer drug research established by the US National Can-
    cer Act of 1971 [23]. The pivotal work of the oncologists Bernard Fisher [24], in the USA, and Gianni Bonadonna [25], in Italy, investigated
    the role of cytotoxic drugs in improving breast cancer cure, inaugurating the concept of adjuvant treatment. In 1975, Gianni Bonadonna
    presented the first report on the efficacy of cyclophosphamide, methotrexate, and fluorouracil (CMF) as adjuvant treatment. These results,
    along with those reported by Dr Fisher´s National Surgical Adjuvant Breast and Bowel Project, raised hopes that chemotherapy could
    have a major role in the management of breast cancer, and were of seminal importance for all the studies on adjuvant systemic therapy
    conducted throughout the world [26]. Since then, breast cancer chemotherapy schedules improved from the methotrexate combinations,
    to anthracyclines and the incorporation of taxanes in the 1990s [27].

    Taking advantage of the available new therapeutic options, visionary physicians started to combine treatment modalities, improving results,
    and reducing morbidities. One of the most preeminent surgeons leading this shift in breast cancer treatment was Umberto Veronesi. Work-
    ing with the oncologist Gianni Bonadonna, at the Istituto Nazionale Tumori, in Milan, the Italian team aimed to perform partial breast surger-
    ies followed by radiotherapy, chemotherapy, and tamoxifen, when applicable. The concept of sentinel lymph node dissection to avoid full
    axillary dissection was also investigated in this context. This less aggressive approach was proved to be as effective as the more radical
    and mutilating Halsted method [28]. Tailoring the surgery method and the adjuvant treatment according to each patient started to be a real-
    ity. The era of personalised therapy was inaugurated.

    Molecular era of cancer treatment

    The proof of concept demonstrated by treatment tailoring according to each patient’s characteristics, staging, and molecular profile was a
    watershed in cancer treatment and research. Advances achieved in breast cancer treatment served as models to improvements in many
    other areas of oncology.

    Conventional histological evaluation of breast cancer by pathologists has identified several histological subtypes; however, this morphol-
    ogy-based breast cancer taxonomy is of limited relevance in terms of tailoring treatment strategies for individual patients. More than 15
    years ago, the employment of the back then newly developed microarrays and the gene expression profiling analysis techniques resulted in
    the identification of the so-called breast cancer intrinsic subtypes [29, 30]. This proved to be a major conceptual breakthrough, since it iden-
    tified different molecular subtypes of breast cancer with distinct clinical behaviour and therapeutic vulnerabilities that can be summarised

    R
    ev
    ie
    w

    4 www.ecancer.org

    ecancer 2017, 11:746

    as follows: (i) luminal A breast cancer with expression profile reminiscent of luminal lineage mammary cells, with indolent clinical behaviour,
    heightened ER-signalling pathways activation and sensitivity to endocrine treatment, (ii) luminal B breast cancer showing poorer prognosis
    as compared to their luminal A counterparts and higher proliferation status coupled with reduced endocrine sensitivity [31], (iii) HER2-like,
    associated with HER2/ErbB2 gene amplification, and (iv) basal-like breast cancer, associated with strong basal epithelial marker expres-
    sion, as well as hormone receptor negativity, high proliferation levels and reduced luminal epithelial and cytokeratin expression [29].

    Treatment and clinical research started to rely not only on morphologic and clinical–pathologic features but on cellular behaviour. Deeper
    understanding of the cellular and molecular biology of breast cancer allowed the detection of genetic aberrations possibly targetable by
    new compounds [32].

    Breast cancer treatment, what next?

    Breast cancer treatment, as it stands today, is selected according to the ‘group of patients’ a specific individual ‘fits in’. As has happened in the
    past, we are coming to a moment where different approaches to breast cancer treatment need to be developed. Selecting treatments according
    to the results of the clinical and available molecular tests, indeed, enriches the chance of response to a given treatment strategy, by classifying
    the patient to a specific subgroup. Nevertheless, individual characteristics that could play a role in the selected patient remain an open area of
    research. Understanding the individual molecular patterns and aberrations of each patient is an approach that could change today’s ‘stratified’
    treatment, based on the intrinsic subtype groups, to a really individualised treatment, based on patients’ specific molecular characteristic.

    The open question is how we take the heritage of knowledge produced by our professors from the past and move on to another level,
    towards really personalised medicine?

    Molecular dissection of breast cancer: intrinsic subtypes and beyond

    During recent years, there has been increasing reference to so-called personalised cancer medicine, defined as: ‘A form of medicine that
    uses information about a person’s genes, proteins, and environment to prevent, diagnose, and treat disease. In cancer, personalised medi-
    cine uses specific information about a person’s tumour to help diagnose, plan treatment, find out how well treatment is working, or make
    a prognosis’ [33]. Personalised medicine has been alternatively described by the term ‘precision medicine’ and it represents an effort to
    individualise the clinical practice applied to any given patient [34].

    In the field of breast cancer, there was an early implementation of some of the above-mentioned principles of personalised cancer medicine,
    namely through the therapeutic targeting of hormone receptor signalling [35]. The successful clinical development of tamoxifen for patients
    with hormone receptor-positive breast cancer represents the archetype of personalised medicine applied in oncology [36]. Subsequently,
    the development of trastuzumab, the first-in-class HER2-blocking agent, for patients with HER2-positive disease exemplified further the
    improved clinical outcomes that personalised cancer medicine can achieve [37]. Of note, in both cases, a true personalisation is yet to be
    achieved; it is more accurate to refer to stratified cancer medicine, given that for individual patients with hormone receptor positive or HER2-
    positive breast cancer treated with endocrine treatment or HER2 blockade, respectively, therapeutic resistance can occur. This indicates
    that further refinement of triaging patients with the respective treatment is needed, for implementing truly personalised cancer medicine.

    The introduction of the intrinsic subtypes of breast cancer, maybe more than anything else, contributed to the dissemination of a conceptual
    advance; that of breast cancer being a group of heterogeneous diseases, associated with different clinical outcomes and therapeutic oppor-
    tunities, as will be detailed later. Both aspects have been capitalised ever since to achieve improved patient stratification in terms of accu-
    rate prognostication and treatment decisions, bringing us several steps closer to the realisation of personalised breast cancer medicine.

    The distinct nature of the breast cancer intrinsic subtypes has been reinforced by studies that coupled gene expression-profiling analysis
    with gene copy number analysis, indicating that the former ones are associated with recurrent copy number aberrations (CNA). More
    recently, the introduction of powerful next-generation sequencing (NGS), also known as massively parallel sequencing, enabled an unprec-
    edentedly detailed characterisation of the molecular underpinnings of breast cancer. Several studies applied this high-throughput molecular
    analysis method to collections of primary breast tumour samples, identifying recurrent gene mutations and/or CNAs among the different
    intrinsic subtypes; some of them are currently pursued as potential therapeutic targets (Table 1) [38, 39].

    R
    ev
    ie
    w

    5 www.ecancer.org

    ecancer 2017, 11:746

    Table 1. Summary of main characteristics of the intrinsic subtypes of breast cancer.
    Luminal A Luminal B HER2-enriched Basal like

    IHC Surrogate ER(+) and/or PR(+),
    HER2(−), Ki67 < 14% (St Gallen)

    ER(+) and/or PR(+),
    HER2(−), Ki67 ≥ 14%
    (St Gallen)

    ER(±), PR(±), HER2(+)
    (St Gallen)

    ER(−), PR(−), HER2(−)
    (St Gallen)

    Prognosis Good Intermediate Poor Poor
    Treatment vulnerability Endocrine treatment Endocrine treatment +

    Cytotoxic chemotherapy
    HER2 blockade Cytotoxic chemotherapy

    Recurrent CNAs:
    Increased copy number

    Decreased copy number

    High-level amplification

    1q, 16p

    16q

    8p11-12,
    11q13-14,
    12q13-14,
    17q11-12,
    17q21-24,
    20q13

    1q, 8q, 17q, 20q

    1p, 8p, 13q, 16q, 17p,
    22q

    8p11-12, 8q, 11q13-14

    1q, 7p, 8q, 16p, 20q

    1p, 8p, 13q, 18q

    17q

    3q, 8q, 10p

    3p, 4p, 4q, 5q, 12q, 13q,
    14q, 15q

    Rare

    Recurrent gene
    mutations (Top-5)

    PIK3CA (45%)
    GATA3 (14%)
    MAP3K1 (13%)
    TP53 (12%)
    CDH1 (9%)

    TP53 (29%)
    PIK3CA (29%)
    GATA3 (15%)
    MLL3 (6%)
    MAP3K1 (5%)

    TP53 (72%)
    PIK3CA (39%)
    MLL3 (7%)
    AFF2 (5%)
    PTPN22 (5%)

    TP53 (80%)
    PIK3CA (9%)
    MLL3 (5%)
    RB1 (4%)
    AFF2 (4%)

    Improving prognostication of patients with early-stage breast cancer

    The advent of microarrays contributed to more accurate breast cancer patient stratification, not only through the identification of the afore-
    mentioned intrinsic subtypes, but also through the development of the so-called first-generation prognostic gene signatures. These multigene
    prognosticators have been developed through the assessment of the epithelial compartment of primary breast tumours. Currently, several of
    these first-generation prognostic gene signatures are clinically available: (i) Endopredict (Sividon Diagnostics, Cologne, Germany) (ii) Mam-
    maPrint (Agendia, Amsterdam, the Netherlands), (iii) MapQuant DX (Ispogen, Marseille, France), (iv) Oncotype DX (Genomic Health, CA,
    USA), (v) PAM50 (Nanostring Technologies, WA, USA), (vi) Theros (bioTheranostics, CA, USA), (v) (Table 2). The importance of such multi-
    gene prognostic data is emphasised in the newest AJCC breast cancer staging system 2017 update. In this update, when molecular data is
    available, it can influence the prognostic classification of the disease, providing additional information to the traditional TNM evaluation [40].

    It should be noted that the above mentioned first-generation prognostic gene signatures can refine the prognostication of patients with
    hormone receptor positive early-stage breast cancer, but not other subtypes of the disease; indeed, the two fundamental biological
    phenomena assessed through them are ER signalling and proliferation status. Furthermore, they disregard the stromal compartment of
    the disease, for which there is increasing evidence supporting its functional importance for malignant progression through interactions
    between cancer and stromal/immune cells [41, 42]. Consecutively, there has been a second wave of efforts trying to further refine the
    prognostication of patients with operable breast cancer, assessing the functionally important stromal component of the disease, as well
    as the differences among the distinct molecular subtypes.

    This has been exemplified by the development of a stroma-derived prognostic predictor (SDPP), a 26-gene prognosticator that was devel-
    oped through an assessment of tumour stroma [43]. Another example of a second-generation prognostic gene signature was developed
    through a comparison of CD10+ cells from cancerous and normal mammary tissues, leading to a 12-gene prognostic signature [44].
    Regarding triple-negative breast cancer, there have been initial efforts to generate immune-related prognostic signatures with promising
    results [45, 46]. More recently, a study mining data from The Cancer Genome Atlas reported two immune/inflammatory gene signatures,
    one associated with poor and one with favourable prognosis among patients with basal-like breast cancer [47].

    R
    ev
    ie
    w

    6 www.ecancer.org

    ecancer 2017, 11:746

    Table 2. Summary of commercially available, first-generation, multigene prognosticators in breast cancer.

    Endopredict MammaPrint MapQuant DX Oncotype DX PAM50 Theros
    Number of Genes
    assessed

    8 70 97 21 50 2

    Types of tumour
    material

    Fresh-frozen FFPE Fresh-frozen FFPE FFPE FFPE

    Technique qRT-PCR DNA microarrays DNA microarrays qRT-PCR nCounter qRT-PCR
    BC subtype
    indication

    ER+/HER2-BC
    treated with
    adjuvant ET

    Stage I or II BC,
    LN-, T ≤ 5 cm

    ER+ Grade 2 BC,
    treated with
    adjuvant ET

    ER+ BC treated
    with adjuvant ET

    and/or CT

    ER+ BC treated
    with adjuvant ET

    ER+ BC treated
    with adjuvant ET

    Prospective
    clinical evidence

    No Yes (MINDACT) No Yes
    (TAILORx)

    No No

    Regulatory
    approval

    No Yes
    (FDA)

    No No No No

    Company Sividon Diagnostics Agendia Ipsogen Genomic Health Nanostring
    Technologies

    bioTheranostics

    Abbreviations: BC: breast cancer, CT: chemotherapy, ER: oestrogen receptor, ET: endocrine therapy, FDA: food and drug administration, HER2: human
    epidermal growth factor receptor 2, LN: lymph node, MINDACT: microarray in node-negative and 1–3 positive lymph node disease may avoid chemotherapy,
    qRT-PCR: q-reverse transcription polymerase chain reaction, TAILORx: trial assigning individualised options for treatment (Rx)

    It should be noted that there is no prospective evidence for second-generation prognostic gene signatures supporting their implementation
    in clinical practice from prospectively conducted randomised studies; thus, their clinical utility still needs to be proven [48]. Truly person-
    alised prognostication of patients with early-stage breast cancer is a goal to be further pursued that could be reached through the assess-
    ment of plasma-based biomarkers.

    Tailoring systemic treatment

    There is an increasing evidence that patient-derived tumour xenografts (PDX) models, alternatively termed tumour avatar models, can be
    valuable tools promoting cancer translational research and advance personalised cancer medicine [49].

    The promise of liquid biopsies

    A conceptual breakthrough has been introduced during recent years in the field of oncology, promising to take us several steps closer to truly
    personalised cancer medicine: that of liquid biopsies [50]. The following types of molecular entities have been reported as possibly relevant
    for oncology: (i) circulating tumour cells (CTCs) that can be assessed at the DNA, RNA and protein level; of note CTCs offer opportunities
    for functional assessment, (ii) cell-free DNA (cfDNA), alternatively called circulating tumour DNA (ctDNA), referring to DNA fragments shed
    to the circulation by cancer cells, (iii) tumour-educated blood platelets, which have been shown to be subjected to changes at the RNA level
    through their interaction with cancer cells [51], and (iv) microvesicles/exosomes, corresponding to extracellular vesicles, carrying protein-
    and nucleic acid-content that corresponds to biological messages transmitted from tumour cells to recipient normal cells [52, 53]. To the
    present day, CTCs and ctDNA are the most extensively studied types of liquid biopsies that could impact the following aspects of oncology:

    A. Cancer screening

    In a study of lung cancer-free subject, CTCs were assessed in patients with chronic obstructive pulmonary disease (COPD) and
    subjects without this condition but matched for smoking habits [54]. There were five patients with COPD (3%) tested positive for

    R
    ev
    ie
    w

    7 www.ecancer.org

    ecancer 2017, 11:746

    CTCs and none from the control group; the annual CT surveillance detected lung parenchyma nodules 1–4 years following the CTCs
    detection, resulting in diagnosis of early-stage lung cancer [54]. Such results have not yet been reported for breast cancer; however,
    it could be envisioned that early CTCs’ detection could complement breast cancer screening for subjects at high risk. Emerging
    powerful technologies of ctDNA detection with increasing sensitivity could be also assessed as potential tools of personalised cancer
    screening [55].

    B. Monitoring of minimal residual disease

    The assessment of minimal residual disease is a well-established clinical practice in the management of patients with leukaemia [56], but
    not in solid tumours; therapeutic decisions in the latter ones are taken on the basis of assessment of the primary tumour and there is no
    measurable parameter to inform about either response to treatment or occurrence of disease recurrence. The assessment of either ctDNA
    or CTCs could be possibly useful for the monitoring of minimal residual disease. In a prospectively conducted study, CTCs were assessed
    using the CellSearch® System evaluated patients with operable breast cancer before the onset of adjuvant chemotherapy administration
    and after chemotherapy [57]. Prior to adjuvant treatment, 21.5% of the patients were tested positive for CTCs, whereas after chemotherapy
    this percentage reached 19.6%; there was no correlation of standard clinico-pathologic risk factors and the detection of CTCs. CTCs
    detection was found to be an independent detrimental prognostic factor, indicating that CTCs could be a measurable parameter assessing
    minimal residual disease in patients with early-stage breast cancer.

    A promising implementation of liquid biopsies would be to guide adjuvant treatment selection and monitor its therapeutic effect; such pre-
    liminary results have been generated through the assessment of disseminated tumour cells (DTCs) in the bone marrow of patients with
    early breast cancer [58].

    C. Guidance of treatment selection

    The ultimate breast cancer medicine personalisation would occur through a ‘real-time’ monitoring of treatment activity, as well as a person-
    alised treatment selection to target therapeutic vulnerabilities at the individual patient level. Theoretically, liquid biopsies could serve both
    purposes. In the classical paradigm, monitoring of treatment activity is performed through conventional image analysis and/or functional
    imaging assessments, complemented by clinical examination, in particular for patients with advanced disease; such objective read-outs to
    monitor treatment activity are lacking for patients with primary disease. This means practically that a patient must receive several cycles
    of treatment prior to the monitoring of the respective effect; for some patients, this means that the futility of a therapeutic strategy will be
    identified only retrospectively.

    Conclusions

    Accumulation of knowledge over time has changed breast cancer from an incurable condition to a range of different diseases with specific
    molecular aberrations, clinical behaviours, and patterns of response to systemic treatments. These improvements are a huge achievement
    for humanity and have been accomplished over time with the contributions of many bright and passionate individuals. To continue this
    legacy, and take it to a new level of excellence, efforts in further understanding breast cancer biology and its interactions with the immune
    system and the microenvironment are being conducted. Prognostic and predictive genetic signatures are already a reality in breast can-
    cer management and are being further refined. Liquid biopsy strategies are in current use for other indications and in research for breast
    cancer, in several settings. Refining our understanding of disease mechanisms and molecular characteristics is key to improving drug
    development and treatment approaches.

    Disclosures

    The authors have declared no conflicts of interest related to the present manuscript.

    R
    ev
    ie
    w

    8 www.ecancer.org

    ecancer 2017, 11:746

    References

    1. van Middendorp JJ, Sanchez GM, Burridge AL (2010) The Edwin Smith papyrus: a clinical reappraisal of the oldest known
    document on spinal injuries Eur Spine J 19(11) 1815–1823 https://doi.org/10.1007/s00586-010-1523-6 PMID: 20697750 PMCID:
    2989268

    2. Allen JP (2005) The art of medicine in ancient Egypt New York : New Haven: Metropolitan Museum of Art ; Yale University Press
    115 p

    3. Papavramidou N, Papavramidis T, Demetriou T (2010) Ancient Greek and Greco–Roman methods in modern surgical treatment
    of cancer Ann Surg Oncol 17(3) 665–667 https://doi.org/10.1245/s10434-009-0886-6 PMID: 20049643 PMCID: 2820670

    4. Sudhakar A (2009) History of cancer, ancient and modern treatment methods J Cancer Sci Ther 1(2) 1–4 https://doi.
    org/10.4172/1948-5956.100000e2

    5. Abu Ali al-Husayn ibn Abdullah ibn Sina (Ibn Sina) (980-1037 CE) [Internet] [cited 2017 Mar 16] Available from: http://www.sciencemu-
    seum.org.uk/broughttolife/people/ibnsina

    6. Abu al-Qasim Al-Zahrawi the Great Surgeon | Muslim Heritage [Internet]. [cited 2017 Mar 16] Available from: http://www.muslimher-
    itage.com/article/abu-al-qasim-al-zahrawi-great-surgeon

    7. Lakhtakia R (2014) A brief history of breast cancer Sultan Qaboos Univ Med J 14(2) e166–e169 PMID: 24790737 PMCID: 3997531

    8. Ghosh SK (2015) Human cadaveric dissection: a historical account from ancient Greece to the modern era Anat Cell Biol 48(3)
    153–169 https://doi.org/10.5115/acb.2015.48.3.153 PMID: 26417475 PMCID: 4582158

    9. Clarke CC (1931) Henri De Mondeville Yale J Biol Med 3(6) 458–481 PMID: 21433498 PMCID: 2606324

    10. Murphy GH (1951) Guy De Chauliac Can Med Assoc J 65(1) 68–71 PMID: 14848801 PMCID: 1821953

    11. Evans CH (2007) John Hunter and the origins of modern orthopaedic research J Orthop Res Off Publ Orthop Res Soc 25(4)
    556–560 https://doi.org/10.1002/jor.20386

    12. Cancer in the Sixteenth to Eighteenth Centuries | American Cancer Society [Internet]. [cited 2017 Mar 16] Available from: https://www.
    cancer.org/cancer/cancer-basics/history-of-cancer/sixteenth-to-eighteenth-centuries.html

    13. Robinson DH, Toledo AH (2012) Historical development of modern anesthesia J Investig Surg Off J Acad Surg Res 25(3) 141–149
    https://doi.org/10.3109/08941939.2012.690328

    14. Brown TM, Fee E (2006) Rudolf Carl Virchow Am J Public Health 96(12) 2104–2105 https://doi.org/10.2105/AJPH.2005.078436
    PMID: 17077410 PMCID: 1698150

    15. Brand RA (2012) Biographical sketch: Charles Hewitt Moore, FRCS (1821-1870) Clin Orthop 470(8) 2075–2076 https://doi.
    org/10.1007/s11999-012-2424-2 PMID: 22695867 PMCID: 3392382

    16. Madden JL, Kandalaft S, Bourque RA (1972) Modified radical mastectomy Ann Surg 175(5) 624–634 https://doi.org/10.1097/00000658-
    197205000-00002 PMID: 4555029 PMCID: 1355229

    17. Banks WM (1882) On free removal of mammary cancer, with extirpation of the axillary glands as a necessary accompaniment
    Br Med J 2(1145) 1138–1141 https://doi.org/10.1136/bmj.2.1145.1138-a PMID: 20750400 PMCID: 2264904

    18. Rankin JS (2006) William Stewart Halsted Ann Surg 243(3) 418–425 https://doi.org/10.1097/01.sla.0000201546.94163.00 PMID:
    16495709 PMCID: 1448951

    19. Sakorafas GH, Safioleas M (2010) Breast cancer surgery: an historical narrative. Part II. 18th and 19th centuries Eur J Cancer
    Care (Engl) 19(1) 6–29 https://doi.org/10.1111/j.1365-2354.2008.01060.x

    https://doi.org/10.1007/s00586-010-1523-6

    http://www.ncbi.nlm.nih.gov/pubmed/20697750

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2989268

    https://doi.org/10.1245/s10434-009-0886-6

    http://www.ncbi.nlm.nih.gov/pubmed/20049643

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2820670

    https://doi.org/10.4172/1948-5956.100000e2

    https://doi.org/10.4172/1948-5956.100000e2

    http://www.sciencemuseum.org.uk/broughttolife/people/ibnsina

    http://www.sciencemuseum.org.uk/broughttolife/people/ibnsina

    http://www.muslimheritage.com/article/abu-al-qasim-al-zahrawi-great-surgeon

    http://www.muslimheritage.com/article/abu-al-qasim-al-zahrawi-great-surgeon

    http://www.ncbi.nlm.nih.gov/pubmed/24790737

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3997531

    https://doi.org/10.5115/acb.2015.48.3.153

    http://www.ncbi.nlm.nih.gov/pubmed/26417475

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4582158

    http://www.ncbi.nlm.nih.gov/pubmed/21433498

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2606324

    http://www.ncbi.nlm.nih.gov/pubmed/14848801

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1821953

    https://doi.org/10.1002/jor.20386

    https://www.cancer.org/cancer/cancer-basics/history-of-cancer/sixteenth-to-eighteenth-centuries.html

    https://www.cancer.org/cancer/cancer-basics/history-of-cancer/sixteenth-to-eighteenth-centuries.html

    https://doi.org/10.3109/08941939.2012.690328

    https://doi.org/10.2105/AJPH.2005.078436

    http://www.ncbi.nlm.nih.gov/pubmed/17077410

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1698150

    https://doi.org/10.1007/s11999-012-2424-2

    https://doi.org/10.1007/s11999-012-2424-2

    http://www.ncbi.nlm.nih.gov/pubmed/22695867

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3392382

    https://doi.org/10.1097/00000658-197205000-00002

    https://doi.org/10.1097/00000658-197205000-00002

    http://www.ncbi.nlm.nih.gov/pubmed/4555029

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1355229

    https://doi.org/10.1136/bmj.2.1145.1138-a

    http://www.ncbi.nlm.nih.gov/pubmed/20750400

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2264904

    https://doi.org/10.1097/01.sla.0000201546.94163.00

    http://www.ncbi.nlm.nih.gov/pubmed/16495709

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1448951

    https://doi.org/10.1111/j.1365-2354.2008.01060.x

    R
    ev
    ie
    w

    9 www.ecancer.org

    ecancer 2017, 11:746

    20. Stockwell S (1983) Classics in oncology George Thomas Beatson, M.D. (1848-1933) CA Cancer J Clin 33(2) 105–121 https://doi.
    org/10.3322/canjclin.33.2.105 PMID: 6402276

    21. O’Malley BW, Khan S (2013) Elwood V. Jensen (1920–2012): father of the nuclear receptors Proc Natl Acad Sci U S A 110(10)
    3707–3708 https://doi.org/10.1073/pnas.1301566110

    22. Goodman LS, Wintrobe MM, and Dameshek W, et al (1984) Landmark article Sept. 21, 1946: nitrogen mustard therapy Use
    of methyl-bis(beta-chloroethyl)amine hydrochloride and tris(beta-chloroethyl)amine hydrochloride for Hodgkin’s disease,
    lymphosarcoma, leukemia and certain allied and miscellaneous disorders ouis S. Goodman, Maxwell M. Wintrobe, William
    Dameshek, Morton J. Goodman, Alfred Gilman and Margaret T. McLennan JAMA 251(17) 2255–2261 https://doi.org/10.1001/
    jama.1984.03340410063036 PMID: 6368885

    23. Vanchieri C (2007) National cancer act: a look back and forward JNCI J Natl Cancer Inst99(5) 342–345 https://doi.org/10.1093/jnci/
    djk119 PMID: 17341721

    24. Wickerham DL, O’Connell MJ, and Costantino JP, et al (2008) The half century of clinical trials of the National Surgical Adjuvant
    Breast and Bowel Project (NSABP) Semin Oncol 35(5) 522–529 https://doi.org/10.1053/j.seminoncol.2008.07.005 PMID: 18929150
    PMCID: 2583142

    25. Ribatti D (2007) The contribution of Gianni Bonadonna to the history of chemotherapy Cancer Chemother Pharmacol 60(3)
    309–312 https://doi.org/10.1007/s00280-006-0410-7 PMID: 17216532

    26. Curigliano G, Valagussa P, and Veronesi U, et al (2016) The influential and inspirational Gianni Bonadonna’s life commitment to
    evidence-based cancer medicine Ann Oncol 27(1) 6–8 https://doi.org/10.1093/annonc/mdv565

    27. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) (2005) Effects of chemotherapy and hormonal therapy for early
    breast cancer on recurrence and 15-year survival: an overview of the randomised trials Lancet Lond Engl 365(9472) 1687–1717
    https://doi.org/10.1016/S0140-6736(05)66544-0

    28. Veronesi U, Banfi A, and Del Vecchio M, et al (1986) Comparison of Halsted mastectomy with quadrantectomy, axillary dissection,
    and radiotherapy in early breast cancer: long-term results Eur J Cancer Clin Oncol 22(9) 1085–1089 https://doi.org/10.1016/0277-
    5379(86)90011-8 PMID: 3536526

    29. Perou CM, Sørlie T, and Eisen MB, et al (2000) Molecular portraits of human breast tumours Nature 406(6797) 747–752 https://
    doi.org/10.1038/35021093 PMID: 10963602

    30. Sørlie T, Perou CM, and Tibshirani R, et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses
    with clinical implications Proc Natl Acad Sci U S A 98(19) 10869–10874 https://doi.org/10.1073/pnas.191367098 PMID: 11553815
    PMCID: 58566

    31. Ades F, Zardavas D, and Bozovic-Spasojevic I, et al (2014) Luminal B breast cancer: molecular characterization, clinical manage-
    ment, and future perspectives J Clin Oncol Off J Am Soc Clin Oncol 32(25) 2794–2803 https://doi.org/10.1200/JCO.2013.54.1870

    32. The TCGA Research Network (2012) Comprehensive molecular portraits of human breast tumours Nature 490(7418) 61–70
    https://doi.org/10.1038/nature11412

    33. Available from: https://www.cancer.gov/publications/dictionaries/cancer-terms?cdrid=561717

    34. Zardavas D, Pugliano L, Piccart M (2013) Personalized therapy for breast cancer: a dream or a reality? Future Oncol Lond Engl
    9(8) 1105–1119 https://doi.org/10.2217/fon.13.57

    35. Tryfonidis K, Zardavas D, and Katzenellenbogen BS, et al (2016) Endocrine treatment in breast cancer: cure, resistance and
    beyond Cancer Treat Rev 50 68–81 https://doi.org/10.1016/j.ctrv.2016.08.008 PMID: 27643748

    36. Jordan VC (2014) Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer Endocr Relat Cancer 21(3) R235–
    R246 https://doi.org/10.1530/ERC-14-0092 PMID: 24659478 PMCID: 4029058

    https://doi.org/10.3322/canjclin.33.2.105

    https://doi.org/10.3322/canjclin.33.2.105

    http://www.ncbi.nlm.nih.gov/pubmed/6402276

    https://doi.org/10.1073/pnas.1301566110

    https://doi.org/10.1001/jama.1984.03340410063036

    https://doi.org/10.1001/jama.1984.03340410063036

    http://www.ncbi.nlm.nih.gov/pubmed/6368885

    https://doi.org/10.1093/jnci/djk119

    https://doi.org/10.1093/jnci/djk119

    http://www.ncbi.nlm.nih.gov/pubmed/17341721

    https://doi.org/10.1053/j.seminoncol.2008.07.005

    http://www.ncbi.nlm.nih.gov/pubmed/18929150

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2583142

    https://doi.org/10.1007/s00280-006-0410-7

    http://www.ncbi.nlm.nih.gov/pubmed/17216532

    https://doi.org/10.1093/annonc/mdv565

    https://doi.org/10.1016/S0140-6736%2805%2966544-0

    https://doi.org/10.1016/0277-5379%2886%2990011-8

    https://doi.org/10.1016/0277-5379%2886%2990011-8

    http://www.ncbi.nlm.nih.gov/pubmed/3536526

    https://doi.org/10.1038/35021093

    https://doi.org/10.1038/35021093

    http://www.ncbi.nlm.nih.gov/pubmed/10963602

    https://doi.org/10.1073/pnas.191367098

    http://www.ncbi.nlm.nih.gov/pubmed/11553815

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC58566

    https://doi.org/10.1200/JCO.2013.54.1870

    https://doi.org/10.1038/nature11412

    https://www.cancer.gov/publications/dictionaries/cancer-terms?cdrid=561717

    https://doi.org/10.2217/fon.13.57

    https://doi.org/10.1016/j.ctrv.2016.08.008

    http://www.ncbi.nlm.nih.gov/pubmed/27643748

    https://doi.org/10.1530/ERC-14-0092

    http://www.ncbi.nlm.nih.gov/pubmed/24659478

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4029058

    R
    ev
    ie
    w

    10 www.ecancer.org

    ecancer 2017, 11:746

    37. Zardavas D, Cameron D, and Krop I, et al (2013) Beyond trastuzumab and lapatinib: new options for HER2-positive breast can-
    cer Am Soc Clin Oncol Educ Book Am Soc Clin Oncol Meet https://doi.org/10.1200/EdBook_AM.2013.33.e2

    38. Shah SP, Roth A, and Goya R, et al (2012) The clonal and mutational evolution spectrum of primary triple-negative breast
    cancers Nature [cited 2017 Mar 16]; Available from: http://www.nature.com/doifinder/10.1038/nature10933 https://doi.org/10.1038/
    nature10933

    39. Zardavas D, Phillips WA, Loi S (2014) PIK3CA mutations in breast cancer: reconciling findings from preclinical and clinical data
    Breast Cancer Res BCR16(1) 201 https://doi.org/10.1186/bcr3605 PMID: 25192370 PMCID: 4054885

    40. Giuliano AE, Connolly JL, and Edge SB, et al (2017) Breast cancer—major changes in the American Joint Committee on Cancer
    eighth edition cancer staging manual CA Cancer J Clin https://doi.org/10.3322/caac.21393

    41. Coussens LM, Zitvogel L, Palucka AK (2013) Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science
    339(6117) 286–291 https://doi.org/10.1126/science.1232227 PMID: 23329041 PMCID: 3591506

    42. Hanahan D, Coussens LM (2012) Accessories to the crime: functions of cells recruited to the tumor microenvironment Cancer
    Cell 21(3) 309–322 https://doi.org/10.1016/j.ccr.2012.02.022 PMID: 22439926

    43. Finak G, Bertos N, and Pepin F, et al (2008) Stromal gene expression predicts clinical outcome in breast cancer Nat Med 14(5)
    518–527 https://doi.org/10.1038/nm1764 PMID: 18438415

    44. Desmedt C, Majjaj S, and Kheddoumi N, et al (2012) Characterization and clinical evaluation of CD10+ stroma cells in the breast
    cancer microenvironment Clin Cancer Res Off J Am Assoc Cancer Res 18(4) 1004–1014 https://doi.org/10.1158/1078-0432.CCR-
    11-0383

    45. Teschendorff AE, Miremadi A, and Pinder SE, et al (2007) An immune response gene expression module identifies a good
    prognosis subtype in estrogen receptor negative breast cancer Genome Biol 8(8) R157 https://doi.org/10.1186/gb-2007-8-8-r157
    PMID: 17683518 PMCID: 2374988

    46. Yau C, Esserman L, and Moore DH, et al (2010) A multigene predictor of metastatic outcome in early stage hormone receptor-
    negative and triple-negative breast cancer Breast Cancer Res BCR 12(5) R85 https://doi.org/10.1186/bcr2753 PMID: 20946665
    PMCID: 3096978

    47. Winslow S, Leandersson K, and Edsjö A, et al (2015) Prognostic stromal gene signatures in breast cancer Breast Cancer Res
    [cited 2017 Mar 10] 17(1). Available from: http://breast-cancer-research.biomedcentral.com/articles/10.1186/s13058-015-0530-2
    https://doi.org/10.1186/s13058-015-0530-2

    48. Michiels S, Ternès N, Rotolo F (2016) Statistical controversies in clinical research: prognostic gene signatures are not (yet)
    useful in clinical practice Ann Oncol 27(12) 2160–2167 https://doi.org/10.1093/annonc/mdw307 PMID: 27634691 PMCID: 5178139

    49. Sia D, Moeini A, and Labgaa I, et al (2015) The future of patient-derived tumor xenografts in cancer treatment Pharmacogenom-
    ics 16(14) 1671–1683 https://doi.org/10.2217/pgs.15.102 PMID: 26402657

    50. Siravegna G, Marsoni S, and Siena S, et al (2017) Integrating liquid biopsies into the management of cancer Nat Rev Clin Oncol
    https://doi.org/10.1038/nrclinonc.2017.14 PMID: 28252003

    51. Best MG, Sol N, and Kooi I, et al (2015) RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and
    molecular pathway cancer diagnostics Cancer Cell 28(5) 666–676 https://doi.org/10.1016/j.ccell.2015.09.018 PMID: 26525104
    PMCID: 4644263

    52. Hannafon BN, Trigoso YD, and Calloway CL, et al (2016) Plasma exosome microRNAs are indicative of breast cancer Breast
    Cancer Res BCR 18(1) 90 https://doi.org/10.1186/s13058-016-0753-x PMID: 27608715 PMCID: 5016889

    https://doi.org/10.1200/EdBook_AM.2013.33.e2

    http://www.nature.com/doifinder/10.1038/nature10933

    https://doi.org/10.1038/nature10933

    https://doi.org/10.1038/nature10933

    https://doi.org/10.1186/bcr3605

    http://www.ncbi.nlm.nih.gov/pubmed/25192370

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4054885

    https://doi.org/10.3322/caac.21393

    https://doi.org/10.1126/science.1232227

    http://www.ncbi.nlm.nih.gov/pubmed/23329041

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3591506

    https://doi.org/10.1016/j.ccr.2012.02.022

    http://www.ncbi.nlm.nih.gov/pubmed/22439926

    https://doi.org/10.1038/nm1764

    http://www.ncbi.nlm.nih.gov/pubmed/18438415

    https://doi.org/10.1158/1078-0432.CCR-11-0383

    https://doi.org/10.1158/1078-0432.CCR-11-0383

    https://doi.org/10.1186/gb-2007-8-8-r157

    http://www.ncbi.nlm.nih.gov/pubmed/17683518

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2374988

    https://doi.org/10.1186/bcr2753

    http://www.ncbi.nlm.nih.gov/pubmed/20946665

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3096978

    http://breast-cancer-research.biomedcentral.com/articles/10.1186/s13058-015-0530-2

    https://doi.org/10.1186/s13058-015-0530-2

    https://doi.org/10.1093/annonc/mdw307

    http://www.ncbi.nlm.nih.gov/pubmed/27634691

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC5178139

    https://doi.org/10.2217/pgs.15.102

    http://www.ncbi.nlm.nih.gov/pubmed/26402657

    https://doi.org/10.1038/nrclinonc.2017.14

    http://www.ncbi.nlm.nih.gov/pubmed/28252003

    https://doi.org/10.1016/j.ccell.2015.09.018

    http://www.ncbi.nlm.nih.gov/pubmed/26525104

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4644263

    https://doi.org/10.1186/s13058-016-0753-x

    http://www.ncbi.nlm.nih.gov/pubmed/27608715

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC5016889

    R
    ev
    ie
    w

    11 www.ecancer.org

    ecancer 2017, 11:746

    53. Skog J, Würdinger T, and van Rijn S, et al (2008) Glioblastoma microvesicles transport RNA and proteins that promote tumour
    growth and provide diagnostic biomarkers Nat Cell Biol 10(12) 1470–1476 https://doi.org/10.1038/ncb1800 PMID: 19011622
    PMCID: 3423894

    54. Ilie M, Hofman V, and Long-Mira E, et al (2014) “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients
    with chronic obstructive pulmonary disease PloS One 9(10) e111597 https://doi.org/10.1371/journal.pone.0111597 PMCID:
    4216113

    55. Lancet oncology (2016) Liquid cancer biopsy: the future of cancer detection? Lancet Oncol 17(2) 123 https://doi.org/10.1016/
    S1470-2045(16)00016-4 PMID: 26868335

    56. Paietta E (2002) Assessing minimal residual disease (MRD) in leukemia: a changing definition and concept? Bone Marrow
    Transplant 29(6) 459–465 https://doi.org/10.1038/sj.bmt.1703388 PMID: 11960263

    57. Rack B, Schindlbeck C, and Jückstock J, et al (2014) Circulating tumor cells predict survival in early average-to-high risk breast
    cancer patients J Natl Cancer Inst 106(5) https://doi.org/10.1093/jnci/dju066 PMID: 24832787 PMCID: 4112925

    58. Naume B, Synnestvedt M, and Falk RS, et al (2014) Clinical outcome with correlation to disseminated tumor cell (DTC) status
    after DTC-guided secondary adjuvant treatment with docetaxel in early breast cancer J Clin Oncol 32(34) 3848–3857 https://doi.
    org/10.1200/JCO.2014.56.9327 PMID: 25366688

    https://doi.org/10.1038/ncb1800

    http://www.ncbi.nlm.nih.gov/pubmed/19011622

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3423894

    https://doi.org/10.1371/journal.pone.0111597

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4216113

    https://doi.org/10.1016/S1470-2045%2816%2900016-4

    https://doi.org/10.1016/S1470-2045%2816%2900016-4

    http://www.ncbi.nlm.nih.gov/pubmed/26868335

    https://doi.org/10.1038/sj.bmt.1703388

    http://www.ncbi.nlm.nih.gov/pubmed/11960263

    https://doi.org/10.1093/jnci/dju066

    http://www.ncbi.nlm.nih.gov/pubmed/24832787

    http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4112925

    https://doi.org/10.1200/JCO.2014.56.9327

    https://doi.org/10.1200/JCO.2014.56.9327

    http://www.ncbi.nlm.nih.gov/pubmed/25366688

    De La Cruz 0

    Claudia De La Cruz

    Donald Moore

    ENC 1143

    Wednesday, February 17, 2021

    A Concise History of Breast Cancer

    Over the past half-century, the understanding of breast cancer biology has transformed, showing us improvements in early techniques, is the most frequently diagnosed type of cancer and second leading cause of cancer death in women after lung cancer. Statistics show that breast cancer affects more than 1,000,000 women worldwide each year, and about 450,000 die from the disease. Breast cancer is a very old disease. The story of breast cancer helps to divide it into three predominant eras times where in the guidelines of care were driven generally by ruling a hypothesis. Breast cancer was treated by surgery or traditional medicine, but rarely cure. Cases described as “breast cancer” were in various civilizations, from Ancient Egypt to the Western Middle Ages. Also, other cases such as tumor progression was also mentioned. Breast cancer was among the most widespread cancers. In the 15th century, researchers and surgeons started to better comprehend the human body. Surgery for breast cancer was implemented, but still was a risky operation at that time since operations were long and painful due to lack of anesthesia and antiseptic conditions. Physician Philippus Theopharstus Aurelus Bombastus von Hohenheim (1493-1541), called Paracelsus, meaning “beyond Celsus”, though that cancer was a product of excess or deficiency of certain fluids rather than an imbalance in the body’s humors. He suggest replace Galen’s black bile by”ens” (entities): ens astrorum (cosmic influences differing with climate and country). In this paper, we will briefly explain the history of breast cancer focusing on the discoveries and inventions in the field of breast cancer detection, analysis, and treatment.

    Greek and Roman period (460 BC-475 AD)

    Prior written history Ancient Greece was know by mythology based on a belief between humans an gods.

    Work Cites

    Ades, Felipe et al. “The past and future of breast cancer treatment-from the papyrus to individualised treatment approaches.” Ecancermedicalscience vol. 11 746. 8 Jun. 2017, doi:10.3332/ecancer.2017.746

    Halyard, Michele Y.; Taghian, Alphonse G. Series: Radiation Medicine Rounds, v. 3, issue 1. New York : Demos Medical. 2012. eBook., Database: 

    eBook Collection (EBSCOhost)

    Lacroix, Marc. Series: Cancer Etiology, Diagnosis and Treatments. New York : Nova Science Publishers, Inc. 2011. eBook., Database: eBook Collection (EBSCOhost)

    Donegan, William L. “History of breast cancer.” Breast cancer (2006): 1-14.

    Reichert FL. Marcus Aurelius Severinus(1580-1656): A Contemporary of Harvey, and Author of the First Work on Comparative Anatomy. Cal. West Med. 1929 Mar;30(3):183-5.

    What Will You Get?

    We provide professional writing services to help you score straight A’s by submitting custom written assignments that mirror your guidelines.

    Premium Quality

    Get result-oriented writing and never worry about grades anymore. We follow the highest quality standards to make sure that you get perfect assignments.

    Experienced Writers

    Our writers have experience in dealing with papers of every educational level. You can surely rely on the expertise of our qualified professionals.

    On-Time Delivery

    Your deadline is our threshold for success and we take it very seriously. We make sure you receive your papers before your predefined time.

    24/7 Customer Support

    Someone from our customer support team is always here to respond to your questions. So, hit us up if you have got any ambiguity or concern.

    Complete Confidentiality

    Sit back and relax while we help you out with writing your papers. We have an ultimate policy for keeping your personal and order-related details a secret.

    Authentic Sources

    We assure you that your document will be thoroughly checked for plagiarism and grammatical errors as we use highly authentic and licit sources.

    Moneyback Guarantee

    Still reluctant about placing an order? Our 100% Moneyback Guarantee backs you up on rare occasions where you aren’t satisfied with the writing.

    Order Tracking

    You don’t have to wait for an update for hours; you can track the progress of your order any time you want. We share the status after each step.

    image

    Areas of Expertise

    Although you can leverage our expertise for any writing task, we have a knack for creating flawless papers for the following document types.

    Areas of Expertise

    Although you can leverage our expertise for any writing task, we have a knack for creating flawless papers for the following document types.

    image

    Trusted Partner of 9650+ Students for Writing

    From brainstorming your paper's outline to perfecting its grammar, we perform every step carefully to make your paper worthy of A grade.

    Preferred Writer

    Hire your preferred writer anytime. Simply specify if you want your preferred expert to write your paper and we’ll make that happen.

    Grammar Check Report

    Get an elaborate and authentic grammar check report with your work to have the grammar goodness sealed in your document.

    One Page Summary

    You can purchase this feature if you want our writers to sum up your paper in the form of a concise and well-articulated summary.

    Plagiarism Report

    You don’t have to worry about plagiarism anymore. Get a plagiarism report to certify the uniqueness of your work.

    Free Features $66FREE

    • Most Qualified Writer $10FREE
    • Plagiarism Scan Report $10FREE
    • Unlimited Revisions $08FREE
    • Paper Formatting $05FREE
    • Cover Page $05FREE
    • Referencing & Bibliography $10FREE
    • Dedicated User Area $08FREE
    • 24/7 Order Tracking $05FREE
    • Periodic Email Alerts $05FREE
    image

    Our Services

    Join us for the best experience while seeking writing assistance in your college life. A good grade is all you need to boost up your academic excellence and we are all about it.

    • On-time Delivery
    • 24/7 Order Tracking
    • Access to Authentic Sources
    Academic Writing

    We create perfect papers according to the guidelines.

    Professional Editing

    We seamlessly edit out errors from your papers.

    Thorough Proofreading

    We thoroughly read your final draft to identify errors.

    image

    Delegate Your Challenging Writing Tasks to Experienced Professionals

    Work with ultimate peace of mind because we ensure that your academic work is our responsibility and your grades are a top concern for us!

    Check Out Our Sample Work

    Dedication. Quality. Commitment. Punctuality

    Categories
    All samples
    Essay (any type)
    Essay (any type)
    The Value of a Nursing Degree
    Undergrad. (yrs 3-4)
    Nursing
    2
    View this sample

    It May Not Be Much, but It’s Honest Work!

    Here is what we have achieved so far. These numbers are evidence that we go the extra mile to make your college journey successful.

    0+

    Happy Clients

    0+

    Words Written This Week

    0+

    Ongoing Orders

    0%

    Customer Satisfaction Rate
    image

    Process as Fine as Brewed Coffee

    We have the most intuitive and minimalistic process so that you can easily place an order. Just follow a few steps to unlock success.

    See How We Helped 9000+ Students Achieve Success

    image

    We Analyze Your Problem and Offer Customized Writing

    We understand your guidelines first before delivering any writing service. You can discuss your writing needs and we will have them evaluated by our dedicated team.

    • Clear elicitation of your requirements.
    • Customized writing as per your needs.

    We Mirror Your Guidelines to Deliver Quality Services

    We write your papers in a standardized way. We complete your work in such a way that it turns out to be a perfect description of your guidelines.

    • Proactive analysis of your writing.
    • Active communication to understand requirements.
    image
    image

    We Handle Your Writing Tasks to Ensure Excellent Grades

    We promise you excellent grades and academic excellence that you always longed for. Our writers stay in touch with you via email.

    • Thorough research and analysis for every order.
    • Deliverance of reliable writing service to improve your grades.
    Place an Order Start Chat Now
    image

    Order your essay today and save 30% with the discount code Happy