Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

707764

research-article2017
TUB0010.1177/1010428317707764Tumor BiologySell

Review
Tumor Biology

Cancer immunotherapy: Breakthrough June 2017: 1­–14 


© The Author(s) 2017
Reprints and permissions:
or “deja vu, all over again”? sagepub.co.uk/journalsPermissions.nav
DOI: 10.1177/1010428317707764
https://doi.org/10.1177/1010428317707764
journals.sagepub.com/home/tub

Stewart Sell

Abstract
From the application of Coley’s toxin in the early 1900s to the present clinical trials using immune checkpoint regulatory
inhibitors, the history of cancer immunotherapy has consisted of extremely high levels of enthusiasm after anecdotal case
reports of enormous success, followed by decreasing levels of enthusiasm as the results of controlled clinical trials are
available. In this review, this pattern will be documented for the various immunotherapeutic approaches over the years.
The sole exception being vaccination against cancer causing viruses, which have already prevented thousands of cancers.
We can only hope that the present high level of enthusiasm for the use of immune stimulation by removal of blocks to
cancer immunity will be more productive than the incremental improvements using previous immunotherapies.

Keywords
Cancer immunotherapy, history of immunotherapy, Coley’s toxins, checkpoint inhibitors, cancer vaccines

Date received: 3 February 2017; accepted: 8 April 2017

Introduction inhibitors) will allow the immune response to individual


cancers to attack and either cure or reduce the growth of
The idea that the immune response could not only protect the cancer. The question remains will this be the long
and defend us against infectious diseases but also provide anticipated breakthrough or will we again see an incre-
a defense against cancer was developed in the late 1800s mental advance in cancer therapy.
with concepts such as the side chain theory and targeted
therapy (magic bullet) by Paul Ehrlich.1 The approached
championed by Ehrlich was applied to cancer chemother- Coley’s toxin
apy as well as to cancer immunology.2 The direction of the The saga of cancer immunotherapy begins in the 1890s
immune response to infectious diseases using vaccines with the development of Coley’s toxin by Dr William B.
(vaccination) has contributed immeasurably to the health Coley described in detail by Stephen Hall in “A commo-
and longevity of humans because of protection not only tion in the blood”7 (see also review by McCarthy8). Shortly
against viral infections3 but also against bacteria,4 fungi,5 after receiving his license to practice medicine in New
and worms.6 As presented in the following, vaccines York in 1890, Coley observed that occasional patients with
against viral infections that cause cancer such as hepatitis inoperable cancers had remissions of their tumors during
B and human papillomaviruses are very effective in pre- life-threatening infections such as severe erysipelas
venting cancers of the liver and cervix (see below). In this
review, many of the attempts to use the immune system
against cancer are presented. These varied and diverse
Wadsworth Center, New York State Department of Health and
approaches first produced striking anecdotal results with Albany College of Pharmacy and Health Sciences, Albany, NY, USA
reports of individual patients with complete cures or sub-
stantial remissions. This pattern has been repeated many Corresponding author:
Stewart Sell, Wadsworth Center, New York State Department of
times with the results of disappointing but incremental Health and Albany College of Pharmacy and Health Sciences, Albany,
advances in the treatment of cancer. There are high expec- NY 12201-2002, USA.
tations that removing blocks to immunity (checkpoint Email: stewart.sell@health.ny.gov

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons
Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial
use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and
Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
2 Tumor Biology

reproduce his results. Because of this failure, the poten-


tially fatal side effects of toxin administration, and the
introduction of irradiation for cancer treatment, the toxin
approach was discontinued. Coley’s positive experience
with early positive results within a limited number of
patients, followed by an inexplicable inability to duplicate
the results, establishes a pattern that besets those working
in cancer immunotherapy to this day (Figure 1). It is likely
that Coley induced what is today known as a cytokine
storm.11 The severe fever and chills are reproduced today
after treatment in some patients, and the specific cytokines
that appear to be involved include IL-2, TNF-β, IL-12,
IFN-α, and TNF-α. Each of these was used in clinical trials
in the 20th century as is reviewed below. The next approach
extends the use of bacterial stimulation to activate the cell-
mediated arm of the immune response to become active
Figure 1.  Projected enthusiasm levels for various cancer
immunotherapies. As early anecdotal reports of spectacular
against cancer.
cures of patient with advanced cancer appear (usually in the
press), there is high enthusiasm, remarkable press coverage, Bacille de Calmette de Guerin
and large amounts of money raised for clinical trials. With
phase I–II clinical trials, the response rate substantially Bacille de Calmette de Guerin (BCG) is an attenuated
decreases and enthusiasm begins to decline, but press reports form of tubercle bacillus used with limited success as a
continue to be high. After phase III trials, a relatively modest vaccine for tuberculosis. A large-scale epidemiologic
response for limited type of cancer is reported (see Table 1).
Finally, because of dangerous side effects and limited long-term
study in Europe suggested a slight decrease in the inci-
remissions, general clinical application is limited. dence of cancer in population immunized with BCG, com-
pared with populations not immunized. This application of
BCG is not recommended in the United States at this time.
(Streptococcus pyogenes). In 1891, he intentionally In the early 1960s, Bast et al.13 at National Institutes of
infected a patient with S. pyogenes, who had a large inop- Health (NIH) demonstrated complete remission of trans-
erable neck tumor. After a life-threatening systemic infec- plantable hepatomas in guinea pigs treated with BCG.
tion, Mr Zola recovered and his tumor showed marked Injection of the tumor cells into the flank led not only to
regression. By 1891 he had treated three patients with local growth of the transplanted tumor but also metastasis
large tumors; all had noticeable reduction in tumor size, to the draining lymph node. Injection of BCG into the pri-
but two of the three patients died.9 During the next few mary transplant site resulted in infiltration of lymphocytes
years, Coley intentionally infected 12 additional cancer around the tumor sites with regression of both the primary
patients: 4 developed severe infections, 2 died, and 2 and metastatic lesions. It was then reported that this
appeared to be cured of their tumors. Eight other patients approach could be used to induce regression of leukemia
did not develop severe infections and tumor reduction was and melanoma lesions in children, but such results were
seen in none of these.10 In 1893, Coley produced tissue again not reproducible. On the one hand, repeated injec-
extracts of cultures of S. pyogenes and Bacillus prodigio- tions of BCG into the bladder of humans with superficial
sus (an endotoxin producing organism) that became known bladder cancer have led to significant remissions.14 This
as Coley’s toxin. Although not known at the time, this application via the bladder produces recruitment of acti-
toxin acts as a superantigen and activates release of vated lymphocytes and macrophages into the bladder wall,
cytokines, including tumor necrosis factor (TNF), interfer- increases ILs in the urine, and induces systemic immunity
ons (IFN), and a number of interleukins (ILs). Treatment to BCG. Long-term remission is achieved in up to 50% of
of a 13-year-old boy with inoperable metastatic tumors by patients; about 35% show no response and 5% have severe
direct injection of the toxin into the tumors resulted in adverse effects.15 Overall, as with Coley’s toxin, the pat-
severe fever and chills and the tumors shrank. This patient tern of response of cancer to BCG did not meet the enthu-
lived to the age of 47 years and died of causes other than siasm of the original reports but at least an incremental
cancer. In 1910 in a lecture at the Royal Society of advance was made by this approach in the treatment of a
Medicine in London, Coley showed photographs of a specific type of tumor, superficial bladder cancer. The non-
patient with a large tumor on the face that became much specific activation of cell-mediated immunity was then
smaller after 63 injections of his toxin.11 By 1914 Coley12 attempted using contact sensitivity haptens that would
had treated over 500 patients, claiming to have induced activate T-cell cytotoxicity to superficial cancer cells
over 150 remissions. However, other physicians could not exposed to the hapten.
Sell 3

Dinitrochlorobenzene TNF was also called cachectin. In other trials, TNF pro-
duced sever systemic effects (cytokine storm) including
Dinitrochlorobenzene (DNCB) acts as a hapten that binds fever, chills, and systemic shock. At the maximum toler-
to tumor cells in the skin and induces a T-cell cytotoxic ated dose, there was little, if any, effect on tumor growth.
response analogous to poison ivy (contact dermatitis). Thus, the prospects for using this very interesting and
Painting superficial skin metastases with DNCB induced a potent factor for treating cancer were not promising.
marked local inflammatory reaction at the lesion sites.16 At In 2006, Ferdy Lejeune et al.24 infused 10× the maxi-
first, it was reported that treatment of the skin lesions was mum tolerated dose of TNF into the isolated limb of a
followed by reduction in the size of internal metastases. patient with multiple melanoma lesions and the lesions
However, this was not reproducible. While DNCB treat- faded away. The systemic shock that was caused by TNF
ment appears to have some cosmetic effect, there is little to that escaped into the systemic circulation had to be aggres-
no evidence to indicate that there is an effect of overall sively treated. Later, the treatment effect was found to be
survival. Treatment of therapy-resistant warts with DNCB enhanced by adding low-dose IFN-γ and chemotherapy
appears to have a beneficial effect.17 If non-specific cell- (Mephalan).25 This was known as “triple therapy.” This
mediated immune did not work, perhaps the effector approach has limited applications because it can only be
cytokines could be used. used on isolated limbs26. Lejeune stated that “You can cure
the limb, but you can’t cure the patient. The current use of
Non-immune-specific cytokines TNF in cancer is in the regional treatment of locally
advanced soft tissue sarcomas and metastatic melanomas
IFN-α or other non-operable tumors in order to avoid amputation
Viral interference, discovered in the 1930s, results in pro- of the limb.27 These results led to further cytokine/cell-
tection of cells or animals against a viral infection, if there mediated approaches.
has been a recent previous infection with a less severe,
unrelated virus. Alex Isaacs and Jean Lindenmann18 found
IL-2 and natural killer cells
that the interference phenomenon could be transferred
from treated cells to uninfected cells by tissue culture In the 1960s, it was observed that supernatants from cultures
fluid. They called the active factor IFN and later showed of lymphocytes stimulated with mitogens, such as phytohe-
that it was produced by the cultured cells and not by the magglutinin (PHA), produced factors that had biological
virus. Later, this was known as IFN-α when Gresser et al.19 effects on other cells (conditioned medium). These factors
in 1969 found a new IFN made by lymphocytes, IFN-γ. In were given names that reflected their biological effects,
1966, Gressner found that IFN-γ not only inhibited virus- such as migration inhibitory factor (MIF) for macrophages,
induced tumors but also reduced growth of epithelial macrophage activating factor (MAF), lymphocyte activat-
tumors. Initial reports of substantial remissions or different ing factor (LAF), and lymphocyte migration inhibitory fac-
cancers were followed by more complete studies that tor (LIF). In 1974, supernatants of PHA were used as
showed little or no effect. In 1986, Jorge Quesada et al.20 at conditioned medium to stimulate growth of cultured leuke-
the MD Anderson Cancer Center treated a 25-year-old mic cells. The stimulatory factor in the conditioned medium
man with advanced hairy cell leukemia with IFN-α. At that was first named LAF and then as T-cell growth factor and
time, there was no effective treatment for this disease. IL-2. In 1986, Steve Rosenberg and Lotze28 at NIH began to
Daily doses of 3 million units achieved complete remis- organize a team to develop immunotherapy of cancer. He
sions. By 1986, IFN-α had been used to treat 30 other started with passive transfer of spleen lymphocytes from
patients with this disease, achieving 9 complete and 17 pigs inoculated with tumor cells from selected patients
partial remissions. IFN-α was subsequently approved for (refer adoptive transfer explained in the following). As no
treatment of hairy cell leukemia by the U.S. Food and positive results were found, he looked for a new approach.
Drug Administration (FDA).21,22 Although not effective Also at NIH, Elizabeth Grimm29 attempted to culture T-cells
for other cancers, the action of this agent reinvigorated the with cancer cells and IL-2 in vitro to obtain a tumor-reactive
field of cytokine treatment of cancer. T-cell line. She did obtain lymphocytes that could kill tumor
cells, but these did not have the properties of T-cells. These
were called lymphokine activated killer cells (LAKs). Many
TNF years earlier, Karl-Eric and Ingegard Hellstrom et al.,30 in
In 1975, Carswell et al.23 found that a novel factor appeared Seattle, had cultured pediatric neuroblastoma cells with
in the serum of mice after treating with BCG and endo- lymphocytes from the same patients to identify immune
toxin had induced necrosis of tumors; they called this sub- cells from the patients that would inhibit growth of the
stance TNF. Another effect of this factor, independently tumor cells (colony inhibition assay). Their preliminary
discovered by others, was to cause wasting disease in findings suggested that some patients with remission had
treated animals with large tumors. Because of this effect, lymphocytes that inhibited colony formation. However, this
4 Tumor Biology

inhibition was significant only when compared to a group of killer cell stimulatory factor (NKSF),37 IL-12 not only
control lymphocyte donors whose lymphocytes had no activates NK cells but also has other actions. These include
effect on colony formation. When other normal donors were activation of CD4+ Th1 helper cells and CD8+ T-cytotoxic
used as a cell source, their lymphocytes did inhibit tumor cells, stimulation of B-cells, inhibition of angiogenesis,
growth even though there was no evidence that they had and blockade of growth of human tumors in immune-defi-
been previously exposed to the tumors. This effect was pro- cient mice, particularly if the IL-12 gene is introduced into
posed to be due to non-immunized normal lymphocytes that fibroblasts.38 In early FDA phase I clinical trials (testing
were named natural killer cells (NKs).31 Grimm29 found that for toxicity and dose level that can be given), no problems
IL-2 exposure increased the activity of NKs, designating were found when patients were primed with low doses of
these cells as LAKs. IL-12 and then given daily doses of IL-12 2 weeks later.
In the meantime, Rosenberg and Michael Lotze28 However, in phase II trials in 1995, higher doses of IL-2
attempted to use large amounts of Il-2 for immunotherapy. were given. Of 15 patients treated with high doses of IL-2,
By 1984, 75 patients had been treated with IL-2 plus LAKs all became seriously ill (cytokine storm) and 2 died. Because
with little or no response. If the dose of IL-2 was increased to of high toxicity, IL-2 is now used in clinical trials as an
the maximum tolerated by the patients, several spectacular addition to gene therapy and only as local injections.39,40
cures were found. However, these patients developed severe The robust response seen in animal studies has not been
side effects, including vascular leak syndrome and life- translated for humans. If one cytokine gave less than satis-
threatening pulmonary edema. These effects were so severe factory results, perhaps more than one cytokine would be
that treatment had to be stopped. Two patients given up for more effective.
dead actually recovered and were judged to be free of cancer.
Biopsies from tissues of successfully treated patients showed
lymphocytic infiltration and necrosis, and over the next
Cytokine mixtures
3 months, the tumor disappeared. In studies of an additional Doses of toxic cytokines, such as IL-2 and TNF, may be
45 patients, a response rate of up to 45% was reported with given in mixtures at lower than toxic levels. An example of
LAK/IL-2 treatment. Again it appeared that a breakthrough such a mixture is a commercially available preparation of
in cancer therapy had been made. Observations by Rosenberg natural cytokines called “leukocyte IL” or “Multikine”
and co-workers received national lay press coverage. When (Cel-Sci Corporation, Vienna, VA, USA) that includes
more complete studies were available, complete responses to IL-2, granulocyte-macrophage colony-stimulating factor
this regimen in 8% of patients and partial responses in (GM-CSF), IFN-γ, and TNF-α.41 Phase II trials suggest
another 10% of patients were found. However, severe side that Multikine may stimulate a healthy immune system.
effects including shock (capillary leak syndrome), chills, However, a phase III clinical study in head and neck can-
fever, nausea, vomiting, diarrhea, cutaneous erythema, ane- cer has been put on hold by the FDA. While cytokines
mia, and moderate-to-severe liver and kidney dysfunction as were being extensively used, there was an increasing inter-
well as thyroiditis cause most centers to discontinue this est and application of transfer of immune active cells.
approach.32,33 As of today, IL-2 is only approved for treat-
ment of mycosis fungoides and Sézary syndrome, tumors of
mature T-helper cells with less than satisfactory results. A Adoptive cell transfer
recent study of these conditions recommends IFNs, reti-
noids, and chemotherapy (doxorubicin and gemcitabine) as
Tumor-infiltrating lymphocytes
well as a monoclonal antibody, alemtuzumab (anti-CD52), In 1894, the preeminent American surgeon at Johns Hopkins,
but not IL-2.34 A more recent approach is the use of IL-2 William Halsted,42 noted that the breast cancer patients
fused with the diphtheria toxin, a chain which delivers the whose tumors had a lymphocytic infiltrate had a better post-
toxin to the IL-2 receptor on the T-helper cell (CD25).35 IL-2 operative prognosis than those patients whose tumor did not
is still used in trials in combination with other therapies such have an infiltrate. He postulated that these lymphocytes
as adoptive transfer of lymphocytes to stimulate T-cell prolif- were reacting against the tumor and were inhibiting growth.
eration (see in the following) in otherwise untreatable cases. However, an alternative hypothesis was presented that the
Overall, these results are very similar to those previously reason that the tumors with infiltrates did not metastasize as
reported for Coley’s toxin, and in fact, it is likely that Coley’s readily as those without was because such tumors required
toxin did produce high levels of IL-2. Now, treatment stimulation by the lymphocytes, and thus, they were more
approaches were then extended to the use of other ILs. dependent on the lymphocytes for their growth than those
without lymphocytes (immune stimulation).43,44 The effect
of lymphocytes on tumors may depend on the type of infil-
IL-12 trating lymphocytes. For example, infiltrates of Th1 cells
IL-12 is produced by activated antigen-presenting cells indicate a good prognosis, whereas infiltrate with Th2 sug-
and has pleiotropic effects.36 Originally called natural gests a poor one.45 In any case, such lymphocytes became
Sell 5

known as tumor-infiltrating lymphocytes (TILs). In 1984, expensive, unless some advance in methodology occurs.
Alexander Knuth et al.46 extracted lymphocytes infiltrating Thus, this approach is an example of impressive biomedi-
a tumor, cultured them with IL-2, and showed that they cal engineering, but it may simply be impractical for more
could lyse tumor cells. These cells were T-cytotoxic cells than occasional use (Figure 1). For more information, see
and were specific for the tumor from which they were iso- the article on the front page of the New York Times (2
lated. Rosenberg at NIH soon reported that reinfusion of August 2016).
TILs back into the patient from induced “substantial remis-
sion” in eight or nine patients with melanoma. After treat-
ment of more patients, a partial response rate of 55% was Immune RNA
claimed,28,47 and in subsequent studies, a much lower The rationale for this approach was that the RNA could be
response was found. This approach has been abandoned. A extracted from immune cells of immunized animals
next step was to transfect the TILs with cytokine genes that (sheep) and used to transfer immunity to cancers to which
might increase the efficiency of TILs. This has not proven the sheep was immunized.55 There are problems with this
effective. A more recent approach being tested is to insert approach. First, if the immune RNA is active, then immu-
chimeric tumor antigen receptors on peripheral blood lym- nity to the normal cells of the patients would most likely be
phocytes cultured from the patient.48 transferred as well. However, if specific tumor immunity
could be induced, the injected RNA would be rapidly
destroyed by the potent ribonucleases present in blood and
Chimeric antigen receptor T-cells
tissue fluids.56 An interesting sidelight of this work is that
In this approach, T-cells from the blood of patients with chest X-rays of lung cancers before and after treatment
refractory B-cell tumors are transfected with anti-CD19, were used to evaluate the size of the tumor seen. However,
so that they recognize essentially all stages of B-cell devel- not only the size of the tumor was smaller but also the size
opment.49,50 Upon transfusion into the patient, these cells of the heart and the density of the ribs were lower.55 Thus,
can effectively attack B-cells. The more recent versions of it has been suggested that the size of the tumor on the chest
these engineered cells contain extracellular domains that X-ray was determined by the extent of radiological expo-
recognize CD19 linked to intracellular activation domains sure. Better success will result through the use of active
which when the receptor binds to CD19 stimulates prolif- stimulation of the immune response to tumor-causing
eration and cytokine production by the transplanted cells. viruses.
They have been used effectively for management of acute
lymphoblastic leukemia and are proposed for use in
Hodgkin’s disease and blast crisis of chronic lymphocytic Tumor vaccines
leukemia. The transfused cells rapidly expand in the
patient.51 This is desirable because the result is that there
Viral vaccines
are more effector cells to attack the cancer. This is undesir- With considerably less fanfare than other immunological
able because these cells also attack normal B-cells, caus- approaches, vaccination against cancers caused by viruses
ing severe clinical complications including B-cell aplasia, has been remarkably successful.57 Thousands, if not mil-
neurologic toxicity, cytokine release syndrome, allergy, lions, of serious cancers have been prevented by this
and anaphylaxis. This has led to consideration of how to approach. This list of viruses and virus-induced cancers
diminish or prevent the side effects.52 An ingenious includes Epstein–Barr virus (Burkett’s lymphoma, etc),
approach is to construct the chimeric receptor T-cell so that hepatitis B virus (HBV, hepatocellular carcinoma (HCC)),
the activation domains of the engineered receptor require a hepatitis C virus (HCV, hepatocellular carcinoma, human
small molecule to join them.53,54 Thus, control of the CAR papillomavirus (cervical cancer, etc.)), Kaposi’s sarcoma
T-cells can be manipulated, so that the activity can be virus, and Merkel cell carcinoma virus.57 Worldwide, over
turned on when necessary. It remains to be seen how much 2 billion people are infected with HBV. HBV infection is
of an impact this approach will eventually have. First, it is believed to be responsible for up to 60% of HCC.58 In
presently applied to a small proportion of leukemia patients endemic areas of HBV infection, introduction of HBV
because about 80% of patients with B-cell cancers respond vaccines has reduced the prevalence of chronic infection in
well with prolonged complete remissions to standard children by more than 90%.59 Thus, vaccination against
chemotherapy and/or monoclonal antibody treatment HBV has already had a significant effect in reduction of
without CAR T-cells (see in the following). Second, the HCC and cirrhosis. There is no doubt that vaccination
serious side effects may eventually prove to be worse than against viruses causing cancers has had more impact on
the treatment for many patients. Third, obtaining periph- preventing cancers than any other immunological
eral blood lymphocytes from each patient, rapidly transfer- approach. If vaccination against tumor-causing viruses
ring them to production laboratories for culture, and works, then maybe vaccination against tumor antigens
genetically engineering the cells are prohibitively would be effective.
6 Tumor Biology

Tumor-specific transplantation antigens


In the 1890s, Paul Ehrlich62 studied transplantation of
tumor cells in animals and noted that growth of meta-
static lesions increased after removal of the primary
tumor. He attempted to generate immunity to cancer by
injection of “weakened” cancer cells. During the early
part of the 20th century, it was found that tumors trans-
planted from one mouse to another were rejected. This,
of course, is because of histocompatibility differences
(graft rejection). In 1943, Ludwig Gross63 observed that
tumors were rejected more slowly in partially inbred
mice, and a decade later, Foley64,65 showed that tumor
grafts growing in inbred mice were rejected by mice
immunized to the tumors. In 1960, George Klein et al.66,67
did the definitive study to show that tumors did have
tumor-specific transplantation antigens (Figure 3).68
Several fundamental concepts were observed from stud-
ies of specific tumor transplantation.

Concomitant immunity.  A tumor bearing individual may be


immune to his/her own tumor cells. Tumor cells taken
Figure 2.  (a) Gross photograph of self-healing malignant from an individual’s own tumor will not grow if injected
melanoma. The growing margins of the cancer are advancing into another site even though the tumor continues to grow
at the same time that the center has “healed.” (b) Lymphocyte at the primary site.69
infiltrate underlying the cancer. (c) Histocytes (macrophages)
containing melanin underlying the “healed” zone. Immunosurveillance.  Newly arising tumors are recognized
by the immune system and held in check by immune
Vaccines against tumor-specific mechanisms.70,71 Of interest, however, there is no increase
in tumor incidence in immunodeficient mice, which can-
antigens not mount an immune response to cancer.72
Self-healing malignant melanoma
Immunoediting (also known as immunoselection). The
The occasional observation of spontaneous remissions of immune systems select for destruction immunogenic
cancer gives rise to speculations of the development of a tumor cells. The result is that the tumors that do arise are
cancer-specific immune response in such patients. Figure 2 weakly antigenic or non-antigenic.73
shows the expanding growth of a self-healing malignant
melanoma, while that the central zone of the tumor shows
healing with no melanoma lesion. Histologically, lympho-
Human tumor-specific vaccines
cyte infiltration of the tumor may be observed along the The first human tumor-specific antigen vaccine was
regressing margins, with collections of melanin-contain- developed after the identification of a tumor antigen on
ing macrophages underlying the normal appearing skin in malignant melanoma cells by Theiry Boon in 1993.74 In
the center of the lesions. Since 1866, there have been 76 1981, Alexander Knuth in Germany sent cancer cells from
reported cases of spontaneous regression of metastases of a patient with multiple metastases to Boon in Belgium.
malignant melanomas.60 The mechanism of these regres- Boon cultured the cells in vitro and treated them with
sions could be immunological, endocrine related, or related mutagens. After irradiation, the non-viable melanoma
to loss of tumor vasculature.61 However, the histological cells were injected back into the patient. After three injec-
findings are consistent with a delayed-type cellular tions of such cells over 3 months, the patient’s tumors dis-
response to the tumor and suggests tumor immunity. Most appeared and with repeated injections did not recur.
studies of tumor-specific immunity in patients address Lymphocytes harvested from the patient very effectively
malignant melanoma or renal cell carcinoma, two tumors killed the cultured tumor cells in vitro. Boon then used
noted for their relatively high incidence of spontaneous IL-2 to stimulate growth of a T-cell line from the patient.
remission of metastases as compared to other cancers. When the T-cell line was tested against a DNA library
Direct evidence for tumor-specific transplantation anti- from tumor cells transfected into tissue-cultured cells, the
gens was originally derived from tumor transplantation cloned T-cell line killed some of the transfected cells.
studies in mice. From one colony of susceptible cells, a 9-amino acid
Sell 7

Figure 3.  Demonstration of tumor-specific transplantation antigens by Geroge Klein et al. (a) Primary methylcholanthrene–
induced cancers from inbred mice were excised and the cells maintained in vitro. (b) When these cells were injected into another
mouse of the same strain, they grew to produce tumors. However, when injected back into mouse A from which the primary
tumor had been removed they did not grow. (c) Then, when the primary mouse was injected with cells from a tumor arising in a
different mouse of the same strain, these cells grew. The conclusion is that the original mouse developed transplantation immunity
to the primary tumor that is specific for that tumor and not for other tumors of the same type: tumor-specific transplantation
immunity.

peptide was identified, which was presented as an antigen Monoclonal antibodies


by class II major histocompatibility complex (MHC).
This peptide was designated as melanoma-associated In 1975, Georges Kohler and Cesar Milstein78 fused anti-
antigen-1 (MAGE-1). Since then, a number of putative body-producing B-cells with cells of multiple myeloma to
tumor antigens have been identified from melanoma, form an immortalized cell line that produced high levels of
prostate, pancreas, and colon tumors.75 Unfortunately, the specific antibody. This Nobel Prize winning discovery
most of these have had very little effect on tumor growth, allowed for the production of large amounts of a single
but some positive effects have been obtained in combina- antibody (monoclonal antibody, -mab). In 1984, Masui
tion with chemotherapy.76 For example, vaccines for et al.79 at University of California, San Diego (UCSD)
­prostate cancer increase survival in metastatic castra- demonstrated that administration of a mouse monoclonal
tion-resistant cancer (Sipuleucel 4.1 months; Prostvac antibody to the human epidermal growth factor receptor
8.5 months).75 Glycoprotein 100 (gp-100) or melanocyte (EGFR) could markedly inhibit the growth of human cancer
protein PMEL is a 661 amino acid transmembrane gly- cells in immunodeficient mice. The mouse model proved
coprotein that is enriched in melanosomes. When applied the principle, but mouse monoclonal antibodies cannot be
topically, it permeates mouse and human skin and used in humans due to their high immunogenicity.
induces T-cell cytotoxic responses.76 It is currently being Subsequently, a massive and incredibly productive effort
tested as an adjunct to chemotherapy in some clinical was made to genetically engineer antibody-producing cells
trials. It appears that high levels of antigen-specific using recombinant DNA to create constructs capable of
T-cells do not necessarily prevent progression of human expression in mammalian cell lines.80 Gene segments
melanomas.77 Thus, although there may be some benefi- capable of producing antibodies were isolated and cloned
cial effects in combination therapies, tumor vaccines do into tissue culture cells that could be grown in a bioreactor.
not play a prominent role in immunotherapy. Paul The antibody proteins produced from the DNA clones
Ehrlich’s1 original hypothesis on the side chain theory were then harvested in large amounts. This was a major
predicted antibodies as the magic bullet for immunother- advance, but even more astounding was that the DNA
apy. This theory is being tested using monoclonal could be engineered to express different parts of the anti-
antibodies. body molecule of mice and humans. Thus, the genetically
8 Tumor Biology

Figure 4.  Genetically engineered monoclonal antibodies. Because mouse monoclonal antibodies cannot be used for human
therapy, various parts of the antibody are replaced by human immunoglobulin chains. These are identified by suffixes: -omab is a
mouse monoclonal; -ximab has human constant heavy and light chains and mouse variable heavy and light chains; zumab (humanized)
has human chains except for mouse complementary binding regions (antibody binding sites); -umab is a fully human antibody.

engineered antibodies may have human constant chains Herceptin (trastuzumab)


and mouse variable chains; it may have human heavy and
light chains, but the complementary determining regions Herceptin is a humanized mouse monoclonal antibody to
(antigen binding sites) of the mouse antibody. In addition, the human epidermal growth factor receptor 2 (HER2)
fully human antibodies have been produced (see Figure 4). present on about 30% of human breast cancers and certain
These products have been coded by specific suffixes. That colon cancers.84 This antibody is used in conjunction with
is, -mab refers to monoclonal antibody, -omab refers to chemotherapeutic regimen of anthracycline, cyclophos-
mouse monoclonal antibody, and -zumab refers to human- phamide, or paclitaxel. Herceptin blocks the cell signaling
ized mouse monoclonal antibodies. necessary for continued growth of the cancer cells. Adding
herceptin to chemotherapy increases disease-free survival
about 10% over chemotherapy alone (84% vs 75%). This
Rituximab is achieved at a cost of increased side effects, such as con-
One of the first anticancer antibodies to be used was the gestive heart failure and acute leukemia.85
human B-cell marker CD20 (rituximab) for treatment of
B-cell malignancies. Rituximab has significantly improved
Other monoclonal antibodies
the treatment of diffuse large-cell B lymphoma and follicu-
lar lymphoma.81 When combined with fludarabine and Overall, over 300 genetically engineered monoclonal
cyclophosphamide, rituximab treatment has achieved long- antibodies have been approved by the FDA for human use
term disease-free survival in chronic lymphocytic leukemia in a variety of diseases in addition to cancer.86 Among the
associated with mutated immunoglobulin heavy chain vari- other cancer-directed monoclonal antibodies are alerntu-
able region.82 About 70% of patients with acute B-cell zumab (targeted at CD53) for chronic lymphocytic leuke-
tumors have complete remissions with the combination of mia, gemtuzumab (directed at CD33) for acute
cyclophosphamide, doxorubicin, vincristine, and pred- myelogenous leukemia, and cetuximab, panitumuab
nisone therapy (CHOP) plus rituximab, whereas the remis- (EGFR), and bevacizumab (vascular endothelial growth
sion rate is 40% when CHOP is used without Rituximab.83 factor (VEGF)) for colon and lung cancer.86 In general,
As is the case in other immunotherapy approaches, success FDA phase II trials showed promise for anti-EGFR thera-
of this regimen is limited to selected cancers. pies, which was not the case in phase III trials. Trials
Sell 9

continue to test this anticancer approach in combination rejection. This effect is not so obvious in human subjects
with chemotherapy. Results of monoclonal antibody treat- who may have decreased Treg activity and lymphocytic
ment of cancer fit the pattern of high initial enthusiasm infiltration of various organs and symptoms of cytokine
and eventual limited effectiveness in clinical trials. The storm.91 Predilection to severe side effects was supported
field of tumor immunity continues to move to new ideas. by findings in the mouse model that included depigmen-
The next idea is that there is an immune response to can- tation and deaths of older mice that were apparently due
cer, but it is inhibited by immune checkpoint regulators. If to autoimmune reactions.92
these checkpoints can be removed or attenuated, then an
individual’s own anticancer response may be able to block
Programmed cell death protein-1/programmed
growth of the tumor.
cell death ligand-1
A second costimulatory immune checkpoint inhibitor is
Immune checkpoint regulation programmed cell death ligand-1 (PD-L1).93 When anti-
inhibitors gen-specific T-cells react with antigen on tumor cells via
the TCR, tumor-derived PD-L1 protein reacts with pro-
Cytotoxic T-lymphocyte-associated protein 4 grammed cell death protein-1 (PD-1) and eliminates
Editorial comments in the scientific and lay press have T-cell killer effects. Like anti-CTLA-4, anti-PD-L1 blocks
recently emphasized breakthrough developments in the this tumor defensive reaction and allows T-cells to attack
very expensive trials of immunotherapy. These articles the cancer cells. Zou et al.94 recently reviewed the results
referred initially to discoveries that immune checkpoint of 22 ongoing clinical trials using variations of this
regulation inhibitors (CPRIs) could be manipulated by approach. The clinical response rate varied from 14.5% in
specific antibodies.87,88 The rationale for this approach is refractory non-small-cell lung cancer to 87% in refractory
that cancers can turn off the immune effector T-cytotoxic Hodgkin’s lymphoma (Table 1). In a comparison with
cells. Specific T-cells are activated by tumor-specific chemotherapy alone, anti-PD-1 (Keytruda) treatment
antigenic peptides presented by dendritic cells to the resulted in an increase of 17% in the 1-year survival rate
T-cell receptor (TCR) and by co-stimulation by reaction of non-small-cell lung cancer.95 Active investigation is
of B7 on the dendritic cells with CD28 on the T-cell.87 underway to predict which patients will respond.96 There
However, IFN-γ production by the T-cell upregulates is good correlation between the immunohistological iden-
production of cytotoxic T-lymphocyte-associated protein tification of high expression of PD-L1 on the cancer cells
4 (CTLA-4) in the cancer cell causing displacement of and clinical response to anti-PD-1.97 Other possible
CD28 on the T-cell. Reaction of the TCR with MHC and response markers are high levels of Th1 cytokines, the
CTLA-4 instead of CD28 with B7 on the dendritic cell presence of tumor antigens,98 lack of immunosuppression,
results in death of the reactive T-cell and protection of the high levels of PD-L1-high cytotoxic T-cells in tumors,99
cancer cell. Treatment with anti-CTLA-4 blocks this low cancer stem cell–like properties,82 and mismatch
inhibition and allows the CD28 to reappear on the T-cell, repair deficiency.98 The largely anecdotal cases of com-
so that it can develop an immune response to the cancer. plete lasting clinical responses have achieved notoriety
An antibody that can accomplish this is ipilimumab. The (New York Times, 31 July, 1 August 2016). Is this a true
preclinical model for this approach was developed by breakthrough or another incremental improvement in can-
Leach et al.,89 who showed that treatment of mice injected cer immunotherapy? It is far from clear. The treatment is
with anti-CTLA-4 greatly inhibited the growth of trans- extremely costly, up to US$250,000 per year per patient.
planted tumor cells. Early trials using anti-CTLA-4 (ipil- This approach has works in only a minority of patients,
imumab) showed that after 3 months of treatment, there and there are potentially severe side effects.100 This side
was increased survival of patients treated with ipili- effect profile is the result of blocking of controls on the
mumab compared to patients treated with tumor antigen immunoreactive T-cells, so that the T-cells also attack nor-
gp 100. Anti-CTLA-4 treatment achieved a plateau of mal organs. As a result, a significant number of patients
survival of about 20% versus none in patients treated successfully treated for cancer develop life-threatening
with gp 100 alone.90 It should be pointed out that these diabetes, cytokine storm, and a variety of autoimmune
were advanced cases for which other therapies had failed diseases.100,101 A major effort is now underway to try to
or showed little effect. In one study, treatment with tras- prevent or limit these untoward reactions. However, they
tuzumab increased responses to CTLA-4 blockade lead- do present serious problems for the general application of
ing to increased T-cell responses in mice with primary CPRI therapies particularly outside of major medical
breast cancers; there was, as well with cytotoxic centers. It would clearly be desirable to target the PD-1/
T-lymphocyte antigen-4 (CTLA-4) blockade, an increase PD-L1 checkpoint exclusively for cancers or non-immu-
in Tregs, which are essential to protect mice from nologically downregulated expression of the PD-L1 gene
immune-mediated autoimmune reactions during tumor in tumor cells.102
10 Tumor Biology

Table 1.  Some examples of response rates in ongoing clinical factor (GM-CSF), and tumor antigens. GM-CSF attracts
trials with anti-PD-1 Nivolumab (Optivo). dendritic cells to the tumor site and activates them. These
Type of cancer Response rate (%) dendritic cells process and present the tumor antigen to
T-cells. The T-cells then identify and destroy the cancer
Advanced melanoma 28 cells throughout the body. This approach is now in FDA
Treatment-refractory melanoma 12.8 phase II clinical trials.
Melanoma without BRAF mutation 40
Non-small-cell lung carcinoma (NSCLC) 14.5
Previously treated advanced NSCLC 17 Therapy directed to the tumor
Advanced NSCLC 20 microenvironment
Relapsed or refractory Hodgkin’s 87
Renal cell carcinoma (RCC) 27 This therapeutic approach is based on the concept that
Previously treated RCC 34 tumor stem cells are protected from chemotherapy by their
Nivolumab (Optivo) followed by ipilimumab   microenvironment. In the case of multiple myeloma, for
 Melanoma 40 example, the myeloma cells form junctions with the bone
  Stage III or IV melanoma 57.6 marrow stromal cells that support the interchange of signal-
ing molecules that protect the myeloma cells. It was postu-
Source: Modified from Thompson et al.82
lated that such junctions could be broken by the proteasome
These are ongoing trials; final conclusive results are not yet available.
Frequently, an early promising complete response is followed by resis- inhibitor bortezomib (PS-341; Velcade) which also pre-
tance to treatment. vents degradation of pro-apoptotic factors and thus allows
programmed cell death.107,108 Treatment of multiple mye-
loma with combinations of bortezomib and thalidomide
Combinatorial therapies were approved by the FDA in 2008. In phase III trials, the
In the last 5 years, the number of clinical trials including combination of bortezomib and melphalan–prednisone is
combinations of chemotherapy, monoclonal antibody, superior to melphalan–prednisolone in the treatment of
tumor antigen, and immunotherapy has increased dramati- older myeloma patients who were not eligible for high-dose
cally and is projected to continue to rise even further.102 In chemotherapy; the complete response rate achieved was
fact, many of the early clinical trials of CPRI therapy were 32%.109 In another study, bortezomib added to other treat-
done on patients whose cancers had resisted conventional ment regimens significantly increased response rates.110,111
chemotherapy.94 The projected potential combinations will There are frequent side effects, such as neuropathy, fevers,
keep clinical trials ongoing for many years. This includes hypoxia, and hypotension. Recently, a humanized mono-
combining chemotherapy and CPRIs with tumor antigen clonal antibody (elotuzumab) to the signaling lymphocytic
(gp-100), adoptive cell transplantation, targeting cancer activation molecule F7 (SLAMF7) receptor has been
stem cells and tumor stroma, microbiota alteration (select- developed.112 This binds to the Fc receptor (CD-16),
ing beneficial bacteria), activation of antigen-presenting activates NK, and blocks interaction between SLAMF7 on
cells, and signal induction inhibitors, such as BRAF and myeloma cells and stromal cells. In preliminary trials, this
MEK inhibitors.103,104 has increased progression-free survival rates by 14% over
lenalidomide plus dexamethasone therapy alone.113
Significant side effects are also associated with this regi-
Oncolytic virus immunotherapy men, including fatigue, diarrhea, and fever.
In this new variation on an old theme of viral oncolysis,
viruses are engineered to infect specific tumors and cause Summary and conclusion
their destruction.105 The FDA approved in 2015 an injecta-
ble form of T-VEC, Imlygic, for the treatment of melanoma The immunotherapeutic approaches to cancer and their
in patients with inoperable tumors.106 T-VEC is a re-engi- proved and projected results are listed in Table 2. The
neered herpes simplex virus 1 (HSV-1), a relatively innocu- results of application of various cancer immunotherapeu-
ous virus that normally causes cold sores. Genetic tics have repeatedly resulted in impressive anecdotal cures
modifications have been made to attenuate the virus (so, it that cause optimism and stimulate hyperbolic press reports.
could no longer cause herpes), to increase selectivity for Decreasing enthusiasm ensues as the results of controlled
cancer cells (so, it destroys cancer cells while leaving clinical trials are reported. Finally, an incremental result is
healthy cells unharmed), and to secrete the cytokine obtained, usually limited to a relatively small number of
GM-CSF (a protein naturally secreted in the body to initiate patients with a highly specific type of cancer. In the case of
an immune response). Inside a healthy cell, the modified Coley’s toxin, the first reported successful immunotherapy,
virus is unable to replicate. Inside a cancer cell, the virus no treatment using this approach currently remains. This is
replicates and produces GM-CSF until cell lysis, releasing due to the inability to reproduce early anecdotal results and
more virus, granulocyte-macrophage colony-stimulating because of important and severe side effects (cytokine
Sell 11

Table 2.  Summary of cancer immunotherapy approaches.


1. Non-specific immunity
  Coley’s toxins (1890–1926) Various cancers Not used
  BCG (1060s) Superficial bladder cancer 50% increase in 5-year survival
  DNCB (1960s) Superficial skin cancer Cosmetic only
  NK/LAK (1970s) Various (melanoma, etc.) Clinical trials ineffective
 IFN-α (1970s) Hairy cell leukemia <50% effective
Tumor necrosis factor Melanoma Isolated limb
 IL-2 Various Stimulate T-cell proliferation
 IL-12 Experimental Too toxic
2. Specific passive immunity (cellular)
  TILs (1980s) Various Discontinued
  Cart-cells (2010) B-cell tumors To be determined
3. Specific passive immunity (humoral)
  Monoclonal antibodies 1980s  
 Alemtuzumab-anti-cd52 Sézary’s syndrome 10% effective
 Herceptin—anti-EGFR Breast, colon 6-month increase in survival
 Rituximab—anti-CD20 B-cell tumors 20% increase in 5-year survival
4. Specific active immunity—vaccination
  Viral (HPV, HBV, HCV, etc.) Various Up to 100% effective
  Melanoma T-CTL peptides Melanoma Anecdotal results
5. Checkpoint inhibitors 2000s
 CTL-4 Various To be determined
 PD-1 Various To be determined
6. Oncolytic virus immunotherapy Experimental
7. Microenvironment
 Bortezomib Multiple myeloma Up to 30% partial response
 Elotuzumab Multiple myeloma To be determined

BCG: Bacille de Calmette de Guerin; DNCB: dinitrochlorobenzene; NK/LAK: natural killer cells/lymphokine activated killer cell; IFN-α: interferon-
α; IL: interleukin; TILs: tumor-infiltrating lymphocytes; EGFR: epidermal growth factor receptor; HPV: human papillomavirus; HBV: hepatitis B virus;
HCV: hepatitis C virus; CTL: cytotoxic T-lymphocyte.

storm). Non-immune-specific approaches including BCG liver, lymphoma, cervical, and other cancers. However,
and IFN-α have resulted in successes in small groups of cancer causing viruses have not been identified for most
patients. Use of IL-2 and IL-2-expanded tumor-infiltrating human cancers. The latest immunotherapeutic emphasis is
lymphocytes was first reported to be a major breakthrough, on checkpoint regulatory inhibitors. Several of these
but like Coley’s toxin did not stand the test of time. The use agents are FDA approved (an anti-CTLA-4 and two PD-1
of adoptive cell transfer is now being tested using geneti- antibodies) for specific oncologic conditions, and multiple
cally engineered chimeric antigen receptor T-cells (CAR-T- additional CPRIs are under development. We are in the
cells) with the receptor linked to cell activation molecules. euphoric phase of the application of these therapies. There
This approach is presently applied to a very small number are many anecdotal cures that have been reported with this
of treatment-resistant B-cell tumors and is enormously approach, but we are still in the mid-phase of larger clini-
expensive as it requires expansion and engineering of lym- cal trials. This is a work in progress and just exactly how
phocytes in vitro from each individual patient. In addition, much overall impact this approach will have remains to be
it is associated with severe long-term side effects. seen. However, the results so far are highly encouraging.
In 1980s, engineered monoclonal antibodies attracted These agents may work in a minority of cancer patients
attention for the treatment of various cancers. Again, after and are associated with severe and long-term side effects,
extensive clinical trials, this treatment is now limited to but in those that respond this is clearly a welcome result.
certain kinds of cancer, for example, herceptin for EGF Hopefully, this approach will prove to be the exception to
receptor-positive breast cancer and anti-CD-20 for B-cell the rule of slight incremental improvement achieved with
tumors. Vaccines using tumor-specific transplantation- other immunotherapies for cancer.
type antigens again have also shown promise for a few
patients, but not as a general application. The most suc- Declaration of conflicting interests
cessful approach that has received less attention is the use The author(s) declared no potential conflicts of interest with
of cancer virus vaccines. These have had an incredible role respect to the research, authorship, and/or publication of this
in preventing and even treating many cancers including article.
12 Tumor Biology

Funding 21. Bonnem EM. Alpha interferon: the potential drug of adju-
vant therapy: past achievements and future challenges. Eur
The author(s) disclosed receipt of the following financial support
J Cancer 1991; 27(Suppl. 4): S2–S6.
for the research, authorship, and/or publication of this article: Dr
22. Vedantham S, Gamliel H and Golumb HM. Mechanism of
Sell’s laboratory is supported by NIH grant CA161694.
interferon action in hairy cell leukemia: a model of effective
cancer biotherapy. Cancer Res 1992; 52: 1056–1066.
References 23. Carswell EA, Old LJ, Kassel RL, et al. An endotoxin-

1. Ehrlich P. Die Wertbemessung ddes Diphterieheilserums induced serum factor that causes necrosis of tumors. Proc
and deren theoretisch Grundlagen. Klin Jahrb 1897; 6: 299– Natl Acad Sci U S A 1975; 72: 3660–3670.
326. 24. Lejeune FJ, Lienard D, Matter M, et al. Efficiency of

2. Strebhardt K and Ulrich A. Paul Ehrlich’s magic bullet concept: recombinant human TNF in human cancer therapy. Cancer
100 years of progress. Nat Rev Cancer 2008; 8: 473–480. Immunol 2006; 6: 6–18.
3. Burton DR. Antibodies, viruses and vaccines. Nat Rev 25. Lienard D, Ewalenko P, Delmotte JJ, et al. High-dose recom-
Immunol 2002; 2: 706–713. binant tumor necrosis factor alpha in combination with inter-
4. Solomon IN and Milner DA. Histopathology of vaccine- feron gamma and melphalan in Isolation perfusion of the limbs
preventable diseases. Histopathology 2017; 70: 109–122. for melanoma and sarcoma. J Clin Oncol 1992; 10: 52–60.
5. Medici NP and Del Poeta M. New insights on the devel- 26. Nooijen PT, Eggermont L, Schalkwijk, et al. Complete

opment of fungal vaccines: from immunity to recent chal- response of melanoma-in-transit metastasis after isolated
lenges. Mem Inst Oswaldo Crus 2015; 110: 966–973. limb perfusion with tumor necrosis factor α and melphalan
6. Hewitson JP and Maizels RM. Vaccination against hel- without massive tumor necrosis: a clinical and histopatho-
minth parasite infection. Expert Rev Vaccines 2014; 13: logical study of the delayed-type reaction pattern. Cancer
473–487. Res 1998; 58: 4880–4997.
7. Hall SS. A commotion in the blood. New York: Henry Holt, 1997. 27. Van Horssen R, Ten Haqen TL and Eggermont AM. TNF-
8. McCarthy EF. The toxins of William B. Coley and the treat- alpha in cancer treatment: molecular insights, antitumor
ment of bone and soft-tissue sarcomas. Iowa Orthop J 2006; effects, and clinical utility. Oncologist 2006; 11: 397–408.
26: 154–158k. 28. Rosenberg S and Lotze MT. Cancer immunotherapy using
9. Coley WB. Contribution to the knowledge of sarcoma. Ann interleukin-2 and interleukin-2-activated lymphocytes.
Surg 1891; 14: 199–220. Annu Rev Immunol 1986; 4: 681–709.
10. Coley WB. The treatment of malignant tumors by repeated 29. Grimm EA. Human lymphokine-activated killer cells (LAK
inoculations of erysipelas: with a report of ten original cells) as a potential immunotherapeutic modality. Biochim
cases. Am J Med Sci 1893; 105: 487–511. Biophys Acta 1986; 865: 267–279.
11. Balkwell F. Tumor necrosis factor and cancer. Nat Rev 30. Hellstrom I, Hellstrom KE, Bill AH, et al. Studies on cel-
Cancer 2009; 9: 351–3719. lular immunity to human neuroblastoma cells. Int J Cancer
12. Coley WB. The treatment of malignant inoperable tumors 1970; 6: 172–178.
with the mixed toxins of erysipelas and Bacillus prodigio- 31. Herberman RB and Holden HT. Natural cell mediated

sus. Brussels: M Weissenbruch, 1914. immunity. Adv Cancer Res 1978; 27: 305–376.
13. Bast RC, Zbar B, Borsos T, et al. BCG and cancer. N Engl J 32. Rosenberg SA, Yang JC, White DE, et al. Durability of
Med 1974; 290: 1413–1420. complete responses in patients with metastatic cancer
14. Lamm DL. Long-term results of intravesical therapy for treated with high-dose interleukin-2: identification of the
superficial bladder cancer. Urol Clin North Am 1992; 19: antigens mediating response. Ann Surg 1998; 228: 307–319.
573–580. 33. Kammula US, White ED and Rosenberg SA. Trends in the
15. Klein E, Holterman OA, Helm F, et al. Topical therapy for safety of high dose bolus interleukin-2 administration in
cutaneous tumors. Transplant Proc 1984; 16: 507–515. patients with metastatic cancer. Cancer 1998; 83: 797–805.
16. Johansson E and Forstrom L. Dinitrochlorbenzene (DNCB) 34. Whittaker S, Hoppe R and Prince HM. How I treat mycosis
treatment of viral warts. A 5-year follow-up study. Acta fungoides and Sézary syndrome. Blood 2016; 127: 3142–3153.
Derm Venereol 1984; 64: 529–533. 35. Wong JK, Mishra A, Hake T, et al. Evolving insights in the
17. Sylvester RJ, Adrian PM and van der Meijden Lamm DL. pathogenesis and therapy of cutaneous T-cell lymphoma
Intravesical bacillus Calmette-Guerin reduces the risk of (Mycosis fungoides and Sezary Syndrome). Br J Haematol
progression in patients with superficial bladder cancer: a 2011; 155: 150–166.
meta-analysis of the published results of randomized clini- 36. Trinchieri G, Gengaraju M, D’Andrea A, et al. Producer
cal trials. J Urol 2002; 168: 1964–1970. cells of interleukin 12. Trends Parasitol 1993; 9: 97–110.
18. Isaacs A and Lindenmann J. Virus interference. I. The inter- 37. Kobayashi M, Fitz L, Ryan M, et al. Identification and puri-
feron. Proc R Soc Lond B Biol Sci 1957; 47: 258–267. fication of natural killer cell stimulatory factor (NKSF), a
19. Gresser I, Bourali C, Levy JP, et al. Increased survival in cytokine with multiple biologic effects on human lympho-
mice inoculated with tumor cells and treated with interferon cytes. J Exp Med 1989; 170: 827–845.
preparations. Proc Natl Acad Sci U S A 1969; 63: 51–57. 38. Tahara H, Zitvogel L, Storkus WJ, et al. Effective eradica-
20. Quesada JR, Hersh EM, Manning J, et al. Treatment of hairy tion of established murine tumors with IL-12 gene therapy
cell leukemia with recombinant alpha-interferon. Blood using a polycistronic retroviral vector. J Immunol 1995;
1986; 68: 493–497. 154: 6466–6474.
Sell 13

39. Lasek W, Radoslaw Z and Jakovisiak M. Interleukin 12: 58. Perz JF, Armstrong GL, Farrington LA, et al. The contribu-
still a promising candidate for tumor immunotherapy? tions of hepatitis B virus and hepatitis C virus infections
Cancer Immunol Immunother 2014; 63: 419–435. to cirrhosis and primary liver cancer worldwide. J Hepatol
40. Tugues S, Burkhard SH, Ohs J, et al. New insights into 2006; 45: 529–538.
IL-12 mediated tumor suppression. Cell Death Differ 2015; 59. Shepard CW, Simard EP, Finelli L, et al. Hepatitis B infec-
2: 237–246. tion: epidemiology and vaccination. Epidemiol Rev 2006;
41. Cell-Sci. Clinical trial status of investigational therapy— 28: 112–125.
Multikine (Leukocyte Interleukin, Injection), http://www.cel-sci. 60. Kalialis LV, Drzesiecki KT and Klyver H. Spontaneous
com/multikine_uses_efficacy_and_clinical_trial_status.html regression of metastases from melanoma: review of the lit-
42. Halsted WS. The results of operations for the cure of cancer erature. Melanoma Res 2009; 19: 275–282.
of the breast performed at the Johns Hopkins Hospital from 61. Wormald RP and Harper JI. Bilateral black hypopyon in a
June, 1889 to January, 1894. Ann Surg 1894; 20: 487–555. patient with self-healing cutaneous malignant melanoma. Br
43. Kaliss N. Dynamics of immunologic enhancement.
J Ophthalmol 1983; 67: 231–235.
Transplant Proc 1970; 2: 59–67. 62. Ehrlich P. The collected papers of Paul Ehrlich (ed F
44. Prehn RT. Immunostimulation of the lymphodependent
Himmelweit). London: Pergamon Press, 1957.
phase of neoplastic growth. J Natl Cancer Inst 1977; 59: 63. Gross L. Intradermal immunization of CSH mice against
1043–1049. a sarcoma that originated in an animal of the same line.
45. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and Cancer Res 1943; 3: 326–333.
location of immune cells within human colorectal tumors 64. Foley EJ. Attempts to induce immunity against mammary
predict clinical outcome. Science 2006; 313: 1960–1964. adenocarcinomas in inbred mice. Cancer Res 1953; 13:
46. Knuth A, Danowski B, Oettgen HF, et al. T-cell-mediated 578–580.
cytotoxicity against autologous malignant melanoma: anal- 65. Foley EJ. Antigenic properties of methylcholanthrene-

ysis with interleukin 2-dependent T-cell cultures. Proc Natl induced tumors in mice of strain of origin. Cancer Res
Acad Sci U S A 1984; 81: 3511–3515. 1953; 13: 835–837.
47. Ettinghausen SE and Rosenberg SA. The adoptive immuno- 66. Klein G, Sjogren HO, Klein E, et al. Demonstration of
therapy of cancer using lymphokine activated killer cells and resistance against methylcholanthrene-induced sarcoma
recombinant interleukin-2. Springer Semin Immunopathol in the primary autochthonous host. Cancer Res 1960; 20:
1986; 9: 51–71. 1561–1572.
48. Cuong CP, Yong CS, Kershaw MH, et al. Cancer immuno- 67. Klein G. Tumor specific transplantation antigens: GHA

therapy utilizing gene-modified T cells: from the bench to Clowes memorial lecture. Cancer Res 1968; 28: 625–635.
the clinic. Mol Immunol 2015; 67: 46–57. 68. Sell S. Immunology of cancer. In: Immunology, immunopathol-
49. Chen R, Song XT and Chen B. CD19 chimeric antigen recep- ogy and immunity. 3rd ed. Ch. 24. Hagerstown, MD: Harper &
tor T cell therapy for the treatment of B cell lineage acute Row, 1980.
lymphoblastic leukemia. Discov Med 2015; 110: 185–190. 69. Gorelik E, Segal S and Feldman M. On the mechanism of tumor
50. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen recep- “concomitant immunity.” Int J Cancer 1981; 27: 847–856.
tor T cells for sustained remissions in leukemia. N Eng J 70. Burnet FM. The concept of immunological surveillance.
Med 2014; 371: 1507–1571. Prog Exp Tumor Res 1970; 13: 1–27.
51. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric 71. Thomas L. Discussion. In: HS Lawrence (ed.) Cellular and
antigen receptors have potent antitumor effects and can humoral aspects of the hypersensitive state. New York:
establish memory in patients with advanced leukemia. Sci Harper & Row, 1970. 20 p.
Transl Med 2011; 3(95): 95ra73. 72. Rygaard J and Poulsen CO. The nude mouse vs the hypoth-
52. Bonifant CL, Jackson HJ, Brentjens RJ, et al. Toxicity and esis of immunological surveillance. Transplant Rev 1976;
management in CAR T-cell therapy. Mol Ther Oncolytics 48: 43–61.
2016; 3: 16011. 73. Bui JD and Schreiber RD. Cancer immunosurveillance,

53. Wu CY, Roybal KT, Puchner EM, et al. Remote control immunoediting and inflammation: independent or interde-
of therapeutic T cells through a small molecular-gated chi- pendent processes? Curr Opin Immunol 2007; 19: 203–208.
meric receptor. Science 2015; 350(6285): aab4077. 74. Boon T. Tumor antigens recognized by cytolytic T lympho-
54. Rodgers DT, Mazagova M, Hampton EN, et al. Switch- cytes. Present perspectives for specific immunotherapy. Int
mediated activation and retargeting of CAR-T-cells for J Cancer 1993; 54: 177–180.
B-cells malignancies. Proc Natl Acad Med U S A 2016; 113: 75. Schlom J. Therapeutic cancer vaccines: current status and
E459–E468. moving forward. J Natl Cancer Inst 2012; 104: 599–613.
55. Pilch YH, Fritze D, Ramming KP, et al. The mediation of 76. Eisenbery F, Machlenkin A, Frankenburg S, et al.

immune responses by I-RNA to animal and human tumor Transcutaneous immunization with hydrophilic recom-
antigens. In: MA Fink (ed.). Immune RNA in neoplasia. binant gp100 protein induces antigen-specific cellular
New York: Academic Press, 1976, p. 149. immune response. Cell Immunol 2010; 266: 98–103.
56. Sell S and Mendelsohn J. Transfer of specific immunity 77. Rosenberg SA, Sherry RM, Morton KE, et al. Tumor pro-
with RNA. Arch Pathol Lab Med 1978; 102: 217–222. gression can occur despite the induction of very high levels
57. Meleif CJ, van Hall T, Arens R, et al. Therapeutic cancer of self/tumor antigen-specific CD8+ T cells in patients with
vaccines. J Clin Invest 2015; 125: 3401–3412. melanoma. J Immunol 2005; 175: 6169–6176.
14 Tumor Biology

78. Kohler G and Milstein C. Continuous cultures of fused cells 97. Daud AI, Loo K, Pauli ML, et al. Tumor immune profiling
secreting antibody of predefined specificity. Nature 1975; predicts response to anti-PD-1 therapy in humans. J Clin
256: 495–497. Invest 2016; 126: 3447–3452.
79. Masui H, Kawamoto T, Sato JD, et al. Growth inhibition 98. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors
of human tumor cells in athymic mice by anti-epidermal with mismatch-repair deficiency. N Eng J Med 2015; 372:
growth factor receptor monoclonal antibodies. Cancer Res 2509–2520.
1984; 44: 1002–1007. 99. Reck M, Rodríguez-Abreu D, Robinson AG, et al.
80. Tansey EM and Catterall PP. Monoclonal antibodies: a wit- Pembrolizumab versus chemotherapy for PD-L1-positive non-
ness seminar in contemporary medical history. Med Hist small-cell lung cancer. N Eng J Med 2016; 375: 1823–1833.
1994; 38: 322–327. 100. Postow M and Wochok J. Toxicities associated with check-
81. Griffin MM and Morley N. Rituximab in the treatment of non- point inhibitor immunotherapy. Release 24.5-C24.268
Hodgkin’s lymphoma—a critical evaluation of randomized Topic 96468. Version 25.0. UpToDate, 2016, https://www.
controlled trials. Expert Opin Biol Ther 2013; 13: 803–811. uptodate.com/contents/toxicities-associated-with-check-
82. Thompson PA, Tam CS, O’Brien SM, et al. Fludarabine, point-inhibitor-immunotherapy
cyclophosphamide, and rituximab treatment achieves long- 101. Nardoo J, Page DB, Li BT, et al. Toxicities of the anti-
term disease-free survival in IGHV-mutated chronic lym- PD-1 and anti-PE-L1 immune checkpoint antibodies. Ann
phocytic leukemia. Blood 2016; 127: 303–309. Oncol 2015; 26: 2375–2386.
83. Huang Y, Ye S, Cao Y, et al. Outcome of R-CHOP or CHOP 102. Lin HY, Chin YT, Nana AW, et al. Actions of I-thyroxine
regimen for germinal center and nongerminal center sub- and nano-daimino-tetrac (Nanoetrac) on PD-L1 in cancer
types of diffuse large B-cell lymphoma of Chinese patients. cells. Steroids 2016; 114: 59–67.
Scientific World J 2012; 2012: 897178. 103. Ledford H. The perfect blend: the next frontier in cancer
84. Volkes EE and Chu E. Anti-EGFR therapies: clinical
immunotherapy lies in combining it with other treatments.
experience in colorectal, lung, and head and neck cancers. Scientists are trying to get the mix just right. Nature 2016;
Oncology 2006; 20(5, Suppl. 2): 15–25. 532: 162–164.
85. Slaman D, Elermann W, Robert N, et al. Adjuvant
104. Ugurel S, Röhmel J, Ascierto PA, et al. Survival of patients
Trastuzumab in HER-2 positive breast cancer. N Engl J Med with advanced metastatic melanoma: the impact of novel
2011; 365: 1273–1283. therapies. Eur J Cancer 2016; 53: 125–134.
86. Campoli M, Ferris R, Ferrone S, et al. Immunotherapy of 105. Kelly E and Russell AJ. History of oncolytic viruses: gen-
malignant disease with tumor antigen-specific monoclonal esis to genetic engineering. Mol Ther 2007; 15: 651–659.
antibodies. Clin Can Res 2010; 16: 11–20. 106. Kaufman HL, Kohlhapp FJ and Zlosa A. Oncolytic viruses:
87. Mellman I, Coukos G and Granoff G. Cancer immunother- a new class of immunotherapy drugs. Nature Rev Drug
apy comes of age. Nature 2011; 480: 480–489. Disc 2015; 14: 642–662.
88. Miller P, Kreuzler Khan T, Thommen DS, et al. Trastuzumab 107. Hideshima T, Bergsaqel PL, Kuehl WM, et al. Advances in
emtansine (TDM1) renders HER2+ breast cancer highly sus- biology of multiple myeloma: clinical applications. Blood
ceptible to CTLA-4/PD-1 blockade. Sci Transl Med 2015; 2004; 104: 607–618.
7(315): 315ra188. 108. Adams J and Kauffman M. Development of the proteas-
89. Leach DR, Krummel MF and Allison J. Enhancement of ome inhibitor Velcade (Bortezomib). Cancer Invest 2004;
antitumor immunity by CTLA-4 Blockade. Science 1996; 22: 304–311.
271: 1734–1736. 109. Reeder CB, Reece DE, Kukreti B, et al. Cyclophosphamide,
90. McDermott S, Lebbé C, Hodi FH, et al. Durable benefit and bortezomib and dexamethasone induction for newly diag-
the potential for long-term survival with immunotherapy in nosed multiple myeloma: high response rates in a phase II
advanced melanoma. Cancer Treat Rev 2014; 40: 1056–1064. clinical trial. Leukemia 2009; 23: 1337–1341.
91. Kuehn HS, Ouyang W, Lo B, et al. Immune dysregulation 110. San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib
in human subjects with heterozygous germline mutations in plus melphalan and prednisone for initial treatment of mul-
CTLA4. Science 2014; 345: 1623–1627. tiple myeloma. N Eng J Med 2008; 359: 906–917.
92. Krummel MF. Quoted by M. Richtel. The New York Times, 111. Harausseau JL, Attal M, Avet-Loiseau H, et al. Bortezomib
4 December 2016. plus dexamethasone is superior to vincristine plus doxoru-
93. Lesokhin AM, Callahan MK, Postow MA, et al. On being bicin plus dexamethasone as induction treatment prior to
less tolerant: enhanced cancer immunosurveillance enabled autologous stem-cell transplantation in newly diagnosed
by targeting checkpoints and agonists of T cell activation. multiple myeloma: results of the IFM 2005-01 phase III
Sci Transl Med 2015; 7(280): 280sr1. trial. J Clin Oncol 2011; 28: 4621–4629.
94. Zou W, Wolchok JD and Chen L. PD-L1 (B7-H1) and 112. Magen H and Muchtar E. Elotuzumab: the first approved
PD-1 pathway blockade for cancer therapy: mechanisms, monoclonal antibody for multiple melanoma treatment.
response, biomarkers, and combinations. Sci Transl Med Ther Adv Hematol 2016; 7: 187–195.
2016; 8: 328sr1. 113. Lonial S, Kaufman J, Reece D, et al. Update on elotu-
95. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the zumab, a novel anti-SLAMF7 monoclonal antibody for
treatment of non-small-cell lung cancer. N Eng J Med 2015; the treatment of multiple myeloma. Expert Opin Biol Ther
372: 2018–2028. 2016; 16: 1291–1301.
96. McGranahan N, Furness AJS, Rosenthal R, et al. Clonal neoan-
tigens elicit T cell immunoreactivity and sensitivity to immune
checkpoint blockade. Science 2016; 351: 1463–1469.

You might also like