Towards Personalized Cancer Therapy

Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1085
Towards Personalized Cancer
Therapy
New Diagnostic Biomarkers and Radiosensitization
Strategies
DIANA SPIEGELBERG
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2015
ISSN 1651-6206
ISBN 978-91-554-9207-6
urn:nbn:se:uu:diva-247539
Dissertation presented at Uppsala University to be publicly examined in Fåhraeussalen,
Rudbecklaboratoriet, Dag Hammarskjölds väg 20, 751 85 Uppsala, Wednesday, 13 May 2015
at 09:15 for the degree of Doctor of Philosophy (Faculty of Medicine). The examination
will be conducted in English. Faculty examiner: Associate Professor Theodosia MainaNock (Molecular Radiopharmacy, INRASTES, National Centre for Scientific Research
“Demokritos”, Aghia Paraskevi Attikis, 15310 Athens).
Abstract
Spiegelberg, D. 2015. Towards Personalized Cancer Therapy. New Diagnostic Biomarkers
and Radiosensitization Strategies. Digital Comprehensive Summaries of Uppsala
Dissertations from the Faculty of Medicine 1085. 62 pp. Uppsala: Acta Universitatis
Upsaliensis. ISBN 978-91-554-9207-6.
This thesis focuses on the evaluation of biomarkers for radio-immunodiagnostics and radioimmunotherapy and on radiosensitization strategies after HSP90 inhibition, as a step towards
more personalized cancer medicine. There is a need to develop new tracers that target cancerspecific biomarkers to improve diagnostic imaging, as well as to combine treatment strategies
to potentiate synergistic effects. Special focus has been on the cell surface molecule CD44
and its oncogenic variants, which were found to exhibit unique expression patterns in head
and neck squamous cell carcinoma (HNSCC). The variant CD44v6 seems to be a promising
target, because it is overexpressed in this cancer type and is associated with radioresistance.
Two new radioconjugates that target CD44v6, namely, the Fab fragment AbD15179 and the
bivalent fragment AbD19384, were investigated with regard to specificity, biodistribution and
imaging performance. Both conjugates were able to efficiently target CD44v6-positive tumors
in vitro and in vivo. PET imaging of CD44v6 with 124I-AbD19384 revealed many advantages
compared with the clinical standard 18F-FDG. Furthermore, the efficacy of the novel HSP90
inhibitor AT13387 and its potential use in combination with radiation treatment were evaluated.
AT13387 proved to be a potent new cancer drug with favorable pharmacokinetics. Synergistic
combination effects at clinically relevant drug and radiation doses are promising for both
radiation dose reduction and minimization of side effects, or for an improved therapeutic
response. The AT13387 investigation indicated that CD44v6 is not dependent on the molecular
chaperone HSP90, and therefore, radio-immunotargeting of CD44v6 in combination with the
HSP90 inhibitor AT13387 might potentiate treatment outcomes. However, EGFR expression
levels did correlate with HSP90 inhibition, and therefore, molecular imaging of EGFR-positive
tumors may be used to assess the treatment response to HSP90 inhibitors.
In conclusion, these results demonstrate how tumor targeting with radiolabeled vectors and
chemotherapeutic compounds can provide more specific and sensitive diagnostic tools and
treatment options, which can lead to customized treatment decisions and a functional diagnosis
that provides more precise and safer drug prescribing, as well as a more effective treatment for
each patient.
Keywords: tumor targeting, radionuclide targeting, HSP90 inhibition, AT13387,
radiosensitization, molecular imaging, combination treatment, EGFR, CD44v6
Diana Spiegelberg, Department of Immunology, Genetics and Pathology, Medical Radiation
Science, Rudbecklaboratoriet, Uppsala University, SE-751 85 Uppsala, Sweden.
© Diana Spiegelberg 2015
ISSN 1651-6206
ISBN 978-91-554-9207-6
urn:nbn:se:uu:diva-247539 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-247539)
I don’t want to believe. I want to know.
Carl Sagan
All of the figures in the introduction have been designed and drawn by
D. Spiegelberg and C. Malmberg.
List of Papers
This thesis is based on the following papers, which are referred to in the text
by their Roman numerals.
I
Spiegelberg, D., Kuku, G., Selvaraju, R., Nestor M. (2014) Characterization of CD44 variant expression in head and neck squamous
cell carcinomas. Tumor Biology, 35(3):2053-62.
II
Haylock, A-K., Spiegelberg, D., Nilvebrant, J., Sandström K., Nestor, M. (2014) In vivo characterization of the novel CD44v6targeting Fab fragment AbD15179 for molecular imaging of squamous cell carcinoma: a dual-isotope study. EJNMMI Research,
6;4(1):11.
III Spiegelberg, D*., Haylock, A-K*., Mortensen, A., Selvaraju, R.,
Nilvebrant, J., Eriksson, O., Tolmachev, V., Nestor, M. Molecular
imaging of CD44v6-expressing squamous cell carcinoma using a
novel engineered bivalent antibody fragment. Submitted manuscript.
IV Spiegelberg, D., Dascalu, A., Mortensen, A., Abramenkovs, A.,
Kuku, G., Nestor, M., Stenerlöw, B. The novel HSP90 inhibitor
AT13387 potentiates radiation effects in squamous cell carcinoma
and adenocarcinoma cells. Submitted manuscript.
V
Spiegelberg, D., Mortensen, A., Selvaraju, R., Eriksson, O., Nestor,
M. Evaluation of biomarkers for imaging and radio-immunotherapy
in combination with HSP90 inhibition in squamous cell carcinomas.
Manuscript.
* Contributed equally
Reprints were made with permission from the copyright holders.
List of Papers not Included in this Thesis
Barta, P., Volkova, M., Dascalu, A., Spiegelberg, D., Trejtnar, F., Andersson, K. (2014) Determination of receptor protein binding site specificity and relative binding strength using a time-resolved competition assay. J Pharmacol Toxicol Methods, 70(2):145-51.
Sahlberg, SH., Spiegelberg, D., Glimelius, B., Stenerlöw. B., Nestor, M.
(2014) Evaluation of cancer stem cell markers CD133, CD44, CD24: association with AKT isoforms and radiation resistance in colon cancer
cells. PLoS One, 9(4):e94621.
Stenberg, J., Spiegelberg, D., Karlsson, H., Nestor, M. (2014) Choice of
labeling and cell line influences interactions between the Fab fragment
AbD15179 and its target antigen CD44v6. Nucl Med Biol, 41(2):140-7.
Sandström, K., Haylock, A-K., Spiegelberg, D., Qvarnström, F., Wester, K., Nestor, M. (2012) A novel CD44v6 targeting antibody fragment
with improved tumor-to-blood ratio. Int J Oncol, 40(5):1525-32.
Spiegelberg, D*., Sahlberg, SH*., Lennartsson, J., Nygren, P., Glimelius, B., Stenerlöw, B. (2012) The effect of a dimeric Affibody molecule
(ZEGFR:1907)2 targeting EGFR in combination with radiation in colon cancer cell lines. Int J Oncol, 40(1):176-84.
Sandström, K., Haylock, A-K., Velikyan, I., Spiegelberg, D., Kareem,
H., Tolmachev, V., Lundqvist, H., Nestor, M. (2011) Improved tumorto-organ ratios of a novel 67Ga-human epidermal growth factor radionuclide conjugate with preadministered antiepidermal growth factor receptor affibody molecules. Cancer Biother Radiopharm, 26(5):593-601.
* Contributed equally
Contents
Introduction................................................................................................... 11
Cancer ...................................................................................................... 11
Detection ............................................................................................. 14
Treatment............................................................................................. 14
Personalized medicine.............................................................................. 15
Tumor targeting........................................................................................ 16
Radio-immunodiagnostics ................................................................... 17
Radio-immunotherapy ......................................................................... 19
Theranostics......................................................................................... 19
Targets and targeting agents .................................................................... 20
Targets of the present study ................................................................. 20
Development of radiolabeled targeting agents .................................... 24
Targeting agents .................................................................................. 24
CD44v6 and EGFR targeting agents ................................................... 26
HSP90 inhibitors ................................................................................. 28
HSP90 inhibition in combination with other treatments .......................... 28
Aim ............................................................................................................... 30
Results........................................................................................................... 31
Paper I New targets for imaging and radio-immunotherapy: CD44 variants ....... 31
Aim and background ........................................................................... 31
Methods ............................................................................................... 32
Results ................................................................................................. 32
Conclusion and discussion .................................................................. 33
Paper II and III CD44v6-targeting fragments for radio-immunodiagnostics .................... 34
Aim and background ........................................................................... 34
Methods ............................................................................................... 34
Results ................................................................................................. 34
Conclusion and discussion .................................................................. 37
Paper IV The HSP90 inhibitor AT13387 potentiates the effects of radiation......... 39
Aim and background ........................................................................... 39
Methods ............................................................................................... 39
Results ................................................................................................. 42
Conclusion and discussion .................................................................. 42
Paper V Molecular imaging in combination with HSP90 inhibition ..................... 44
Aim and background ........................................................................... 44
Methods ............................................................................................... 44
Results ................................................................................................. 45
Conclusion and discussion .................................................................. 46
Concluding remarks ...................................................................................... 49
Future studies ................................................................................................ 51
Acknowledgements....................................................................................... 53
References..................................................................................................... 56
Abbreviations
17-AAG
17-DMAG
ALK
AKT
AT13387
ATM
bFGF
BIWA
CAT
CD
CD44s
CDv
cmAb
CPS
CSC
CT
Da
DARPins
DNA
DNA-PKcs
DSB
EGF
EGFR
ErbB
ERK
Fab
FACS
FAK
FAR
FBS
FDA
FDG
17-Allyl-17-Demethoxygeldanamycin
17-(Dimethylaminoethylamino)-17demethoxygeldanamycin
Anaplastic lymphoma kinase
V-akt murine thymoma viral oncogene homolog
2,4-dihydroxy-5-isopropyl-phenyl-[5-(4-methylpiperazin-1-ylmethyl)-1,3-dihydro-isoindol-2-yl]
thanone, l-lactic acid salt
Ataxia telangiectasia mutated
Bovine fibroblast growth factor
Bivatuzumab
Chloramine T
Cluster of differentiation
Standard CD44
CD exon variant
Chimeric monoclonal antibody
Counts per second
Cancer stem cell
Computed tomography
Dalton
Designed ankyrin repeat proteins
Deoxyribonucleic acid
DNA-dependent protein kinase catalytic subunit
Double-strand break
Epidermal growth factor
Epidermal growth factor receptor
Erythroblastic leukemia viral oncogene homolog
Extracellular signal regulated kinase
Antigen binding fragment
Fluorescent-activated cell sorting
Focal adhesion kinase
Fraction of activity released
Fetal bovine serum
United States Food and Drug Administration
Fluorodeoxyglucose
GIST
Gy
HER2, 3, 4
HOP
HSP
HNSCC
HPV
LET
mAb
MET
MIP
mM
MRI
MTT
Na(TI)
nM
NRG1, 2, 3, 4
IC50
%ID/g
IgG
IHC
iodogen
i.v.
p53
PET
PFGE
p.i.
PTK
RECIST
scFvs
SD
SEM
SPECT
SPR
TGFα
TKI
VEGFR
µM
Gastrointestinal stromal tumors
Gray
Human EGFR 2, 3, 4
HSP-organizing protein
Heat shock protein
Head and neck squamous cell carcinoma
Human papilloma virus
Linear energy transfer
Monoclonal antibody
Hepatocyte growth factor receptor
Maximum intensity projection
Millimolar
Magnetic resonance imaging
(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium
bromide
Sodium iodide (activated with thallium)
Nanomolar
neuregulins 1, 2, 3, 4
Median inhibitory concentration
Percentage of the injected dose measured per gram of
tissue or organ
Immunoglobulin, class G
Immunohistochemistry
1,3,4,6-Tetrachloro-3α,6α-diphenylglycouril
Intravenous injection
Tumor protein 53
Positron emission tomography
Pulsed-field gel electrophoresis
Post injection
Protein tyrosine kinase
Response evaluation criteria in solid tumors
Single-chain antibody variable fragments
Standard deviation
Standard error of the mean
Single-photon emission tomography
Surface plasmon resonance
Transforming growth factor alpha
Tyrosine kinase inhibitor
Vascular endothelial growth factor receptor
Micromolar
Introduction
Cancer
Statistically, more than 1 in 9 of everyone reading this text will die due to
cancer, and virtually everyone will come into close contact with the disease
in their lifetime through family and friends [1]. It is not surprising that
fighting cancer, or finding “the cure for cancer”, is considered by many as
the holy grail of biomedical research - the very epitome of our struggles as
medical scientists.
However, cancer is not a single disease. Rather, the term cancer summarizes a very broad class of diseases defined by a common mechanism: loss of
control of the own cells of the affected organism, which involves uncontrolled cell proliferation, invasion into normal unaffected tissues and the
capability for the development of metastases in other parts of the body. Over
100 different types of cancer exist, with an overwhelmingly diverse flora of
subtypes and molecular mechanisms. It is clear that the probability of a single, universal cancer cure is exceedingly small.
Cancer has been with us since the dawn of multicellular organisms, and
throughout human history. Relatively recently, cancer has emerged as a
leading cause of death in the developed world, following the near elimination of infectious and parasitic diseases, malnourishment, and the “neglected
diseases of the developing world”: homicides, suicides, and war. The same
transition is expected to increase the cancer mortality in the developing
world in the following decades, as the people there progress into transitional
or developed societies. In 2008, 7.6 million people (approximately 13 % of
all recorded deaths this year) died due to cancer, and deaths from cancer
worldwide are projected to continue to rise to over 13.1 million in 2030 due
to this shift [1]. The economic impact of cancer on both the healthcare system and society as a whole is severe; in 2008, it was estimated to be 201.5
billion US dollars in the USA alone. This figure includes both direct treatment costs (77.4 billion) and indirect costs to society as a result of lost
productivity due to premature death (124 billion). The great cost in human
life as well as economic resources makes cancer treatment a highly important field of research [2, 3].
Most instances of cancer derive from a single abnormal cell. These abnormalities can be caused randomly by mistakes in DNA replication, by
exposure to carcinogens such as radiation or a plethora of carcinogenic
11
chemical substances, by viruses or by inherited genetic disorders [2-4]. The
transformation from normal cells to cancer cells is generally attributable to
mutations or alterations in two different groups of genes: activation of oncogenes and inactivation of tumor suppressor genes. Still, a single mutation is
not sufficient to cause cancer; typically the affected cells gradually accumulate mutations over time and spiral increasingly out of control. This process
is constantly ongoing, but in the vast majority of cases, the abnormal cells
are swiftly cleared by the immune system or undergo apoptosis. Eventually
however, one abnormality will escape the multiple layers of protective systems, which is why the cancer risk increases with age. The first step in this
process is an abnormal proliferation of tissue, called a neoplasm, which typically forms a tumor. There are three general forms of tumors that can be
distinguished: benign, pre-malignant (carcinoma in situ) or malignant. Benign tumors are not cancerous; they do not grow in an infinite or aggressive
way, nor do they invade other parts of the body. Nevertheless they can be
dangerous, e.g., if the tumor imposes pressure upon vital organs, particularly
the brain. The term pre-malignant tumor refers to a pre-cancerous condition
defined by the absence of invasion of surrounding tissues, which seems to be
the interstate between benign and malignant tumors. If pre-malignant tumors
remain untreated the condition can lead to malignant growth. Malignancies
or malignant tumor growth are generally referred to as cancer. These tumors
invade and destroy the surrounding tissue and may potentially spread to
other parts of the body to form metastases; at this stage of the disease the
cancer is life-threatening, and the likelihood of successful treatment is very
low [5-7].
It is important to understand that tumors are highly heterogeneous, and
there is evidence that a large proportion of tumors consists of relatively inactive and sensitive bulk cells together with small populations of more aggressive tumorigenic cells. Interestingly, these subpopulations also seem to be
more resistant to many cancer drugs and to possess an elevated tolerance to
radiation, indicating that they are more important for treatment outcomes.
They are often called cancer initiating cells or “cancer stem cells” (CSC),
due to their functional similarities to the stem cells of the body: the capability to undergo self-renewal and differentiation [8].
This study focuses on two cancer types, originating from the head and
neck and the colorectal area, which are presented in the following section.
Head and neck squamous cell carcinoma
Head and neck cancer comprises tumors of diverse origin, such as the oral
cavity, sinonasal cavity, salivary glands, pharynx, larynx and lymph nodes in
the neck. More than 95 % of head and neck cancers are squamous cell carcinomas (HNSCC) rising from the epithelial cells of in these regions [9].
HNSCC represents the fifth most common solid cancer worldwide, with
12
approximately 0.5 million new cases diagnosed every year [1]. Tobacco and
alcohol use, as well as infection with high-risk types of human papilloma
virus (HPV), are important risk factors for head and neck cancers [9]. Most
patients with HNSCC present with advanced-stage locoregional disease with
metastases that are located primarily in regional lymph nodes in the neck
area. Current treatment options consist of multiple-modality therapy with
surgery, radiation, and chemotherapy. Despite significant improvements in
these modalities, the overall 5-year survival rates are less than 50 %, a figure
that has remained relatively unchanged for the past 30 years [10]. This indicates a need for earlier diagnosis and additional treatment options that target
the disease more effectively and with reduced toxicity. Several aberrant signaling pathways and abnormal expression oncogenes and oncoproteins have
been identified in HNSCCs, including CD44v6, epidermal growth factor
receptor (EGFR) and heat shock 90 protein 90 (HSP90) [9-12], which are
described later in the section Targets of the present study.
Colorectal adenocarcinoma
After lung and breast cancer, colorectal cancer is the third most frequent
type of cancer worldwide and the second cause of all cancer deaths, with
approximately 1 million newly diagnosed cases every year [1, 13]. Colorectal cancer occurs in the colon, rectum and/or cecum with appendix. At diagnosis many patients have already developed metastases in the liver or lung,
which is one reason why colorectal cancer is associated with a poor prognosis in late stages and has a 5-year survival rate of less than 10 % [14]. Because the progression is quite slow, the risk of acquiring colorectal cancer
increases with age; most patients are 50 years or older. Other factors that
increase the risk for developing colorectal cancer are related to the general
lifestyle: for example, an unbalanced diet that is high in fat and with a high
intake of red meat, alcohol and small amounts of fresh fruits and vegetables,
together with sparse exercise [15].
Multiple signaling pathways and constitutively activated signaling proteins are involved in the pathogenesis of colorectal cancer, e.g., APC, PTEN,
KRAS, BRAF, PI3K and NRAS [16]. KRAS, BRAF and NRAS mutations
can all activate the RAS/RAF/MAP kinase pathway, which is located downstream of growth factor receptors such as EGFR. EGFR plays a key role in
the activation of these pathways and is commonly overexpressed in metastatic colorectal cancer [17, 18]. Furthermore, metastatic progressing of colorectal cancer is initiated by cancer cells expressing CD44v6, which can
function both as a biomarker and as a therapeutic target [19].
13
Detection
The early detection, diagnosis and treatment of cancer increases the patient’s
chance for a full recovery. Cancer is most commonly discovered during a
physical exam or as a result of routine tests. Detection methods include endoscopy of the neck, esophagus and gastrointestinal tract, histological analysis of tissue samples (biopsies), laboratory tests of body fluids including
blood and urine as well as various imaging techniques. Imaging methods
that are in clinical practice include X-ray, ultrasound, magnetic resonance
imaging (MRI), computed tomography (CT) and radionuclide based techniques such as single-photon emission tomography (SPECT) and positron
emission tomography (PET) [20]. Molecular imaging using SPECT and PET
is further described in the Radio-immunodiagnostics section.
Treatment
Alternatives for the treatment of cancer depend on several factors, such as
the type and stage of the cancer, location, whether the cancer has recurred
subsequent to earlier treatment, and the general health condition of the patient. However, the most common treatment is surgical removal of the tumor
together with chemotherapy and/or radiation therapy, and/or support provided by biologic therapy after surgery [6, 21].
Radiation therapy
At present, treatment with ionizing radiation is one of the most successful
treatment modalities for solid cancers and is applied to over 50 % of all cancers at one stage of their management [3, 22]. Radiation in the form of high
energy X-rays, γ-rays and charged particles is used for adjuvant, neoadjuvant or palliative therapy to treat nearly all types of cancer. The common mechanism of action is ionization of molecules in the cells. Although
all molecular constituents of the cell are affected, the desired effect of applied radiation is DNA damage. The rays/particles can either directly ionize
the DNA molecule, or more likely, indirectly affect it via the formation of
free hydroxyl radicals through the ionization of water molecules. The induced effects are manifested as base damage and single or double-strand
breaks (DSB) in the DNA [22]. The goal is to disrupt the genetic material to
such an extent that repair and replication cannot proceed, thus inactivating or
killing the cell.
However, radiation induces DNA breakage in both normal and cancer
cells. To some extent this damage can be mitigated by moving and focusing
the beam to minimize the exposure to surrounding tissue. Furthermore, most
cancer cells have lost or have deficient apoptotic or cell cycle control, as
well as poor or lack of DNA repair responses. When normal cells with a
functioning cell cycle checkpoint system are affected by DNA damage the
14
cell cycle phase does not transition to allow the repair of the breaks. The
rapid growth and abnormal cell cycle of tumor cells instead sensitizes them
to radiation. DNA damage is not corrected, and errors are inherited through
subsequent cell divisions, resulting in accumulating damage to the cancer
cells and finally leading to reduced growth and cell death [23].
Chemotherapy
Drug therapies, which are applied to reduce tumor size and growth, as well
as to suppress both the development and spread of metastases, are referred to
as chemotherapy. Chemotherapy treatment can be used as an adjuvant,
meaning after surgical intervention, or as a neo-adjuvant before surgery.
Chemotherapy is even administered as the primary therapy for palliative
treatment to reduce the severity of symptoms and disease progression. The
application depends on the progression of the cancer; for example, an adjuvant treatment is usually only given if the cancer has started to spread to
lymph nodes or formed metastases in other tissues [6]. Many drugs used in
chemotherapy are directed against the growth and division of cells in the
entire organism, with effects similar to radiation therapy but generally without the targeting possibilities, which greatly increases the risk for side effects [6]. Thus, the concept of personalized medicine and targeted therapy
has become increasingly clinical relevant.
Personalized medicine
The concept of personalized cancer medicine is based on customized treatment decisions after functional diagnosis. Because cancerous diseases are
very heterogeneous, not all patients benefit from the same treatment. Patients with a different genetic background may only partially respond or not
respond at all to the treatment, or in the worst cases, they may develop adverse effects. Therefore, it is important to tailor healthcare to the individual
characteristics, needs, and preferences of the patient. The growing understanding of molecular mechanisms and genetics provides more precise diagnoses, safer drug prescribing, and more effective personalized treatment.
Ideally, personalized medicine is practiced during all stages of disease, including prevention, diagnosis, therapy, and follow-up [24-26].
The concept of personalized cancer medicine is gradually integrated in
the clinical practice, and during the last decade, more than 50 new targeted
drugs for the treatment of solid and hematological malignancies have been
approved by the FDA [26]. However, personalized medicine is not yet a
commonly used cancer treatment strategy, potentially due to the lack of experience, relatively high costs of the treatments and the accompanying diagnoses [27]. The concept of personalized medicine is presented in Figure 1.
15
Figure 1. The concept of personalized medicine. Molecular diagnostic tools are
used to match patients with treatments that provide them with the best outcomes,
while traditionally treated patients with the same diagnosis do not necessarily benefit from the same treatment.
Tumor targeting
Tumor targeting relies on the usage of highly specific targeting agents, such
as monoclonal antibodies (mAb), which can target specific molecular abnormalities in the tissue of concern. These targets are antigens, which are
specific or overexpressed on tumor cells but are absent or expressed at a low
level on normal cells, to avoid damaging the healthy tissue. This concept can
be utilized for diagnostic imaging and therapy purposes. The cancer cells
either can be selectively destroyed by the targeting agents themselves when
they are also designed as effectors, or a payload of cytotoxic substances can
be delivered to the malignancy with a lower risk of affecting healthy tissues.
This strategy of delivery is used, e.g., in radio-immunotargeting, in which a
16
Figure 2. The concept of radio-immunotargeting. A radiolabeled compound binds to
a target that is overexpressed on the surface of tumor cells, while normal cells do
not (or to very small extent) express that target.
radionuclide is attached to the targeting agent [23] (e.g., to an antibody), as
demonstrated in Figure 2. The idea behind this setup is that a coupled radioactive emitter can be detected using PET, SPECT or a gamma camera for
imaging purposes. Another application is the use of a therapeutic radionuclide coupled to the targeting agent. This tracer can selectively destroy cancer cells with fewer of negative side effects compared with conventional
radiotherapy [28].
A quite different principle of targeted treatment is the usage of drugs that
affect cell-specific pathways and signaling. In this scenario, the idea is to
selectively adjust the cellular behavior of cancer cells by affecting the signaling pathways, for example by blocking or degrading constitutive activated signaling proteins that cause abnormal proliferation. The drug AT13387
which is examined in Papers IV and V uses this principle to target HSP90.
New potential targets for radio-immunotherapy and diagnostic imaging,
namely EGFR and CD44 variants, are evaluated in Papers I-III.
Radio-immunodiagnostics
Radio-immunodiagnostics aim to non-invasively visualize, characterize and
quantify the molecular interactions of biological processes in real time; in
living cells, tissues and intact subjects [20, 29]. The wide range of applications of molecular imaging includes diagnostics, drug discovery and devel17
opment, theranostics and personalized medicine. It can be used to precisely
locate diseased cells in the body, which allows for appropriate treatment
planning and treatment response monitoring [29]. Radio-immunodiagnostic
imaging techniques are most useful in combination with CT or MRI scans,
which allow co-localization of the tracer and the precise anatomical position,
so called “multimodality imaging” [20, 29]. Table 1 summarizes the features
of the central imaging modalities that are used preclinically and clinically.
Molecules that are suitable for imaging are e.g., antibodies, antibody
fragments, scaffold proteins, ligands and peptides. Small molecules (below
100 kDa) are preferable for imaging since they provide high contrast images
in shorter time intervals due to better tissue penetration, shorter circulation
and biological half-lives and efficient clearance from the body [29]. Imaging
vectors are further described in the section Targeting agents.
Table 1. Features of the central imaging modalities used preclinically and clinically.
– low, + moderate, ++ high costs.
Modality Temporal Spatial Resolution
Resolution
Sensitivity
Cost
Safety Profile
CT
Minutes
ND
+
Ionizing radiation
MRI
Minuteshours
Minutes
10-3-10-5 M
++
10-10-10-11 M
+
No ionizing
radiation
Ionizing radiation
10-11-10-12 M
++
Ionizing radiation
SPECT
PET
Secondsminutes
50-200 µm (preclinical)
0.5-1 mm (clinical)
25-100 µm (preclinical)
∼1 mm (clinical)
1-2 mm (preclinical)
8-10 mm (clinical)
1-2 mm (preclinical)
5-7 mm (clinical)
Single-photon emission computer tomography (SPECT)
In SPECT, tracers labeled with radionuclides (e.g., 99mTc, 111In, 123I, 67Ga)
that emit gamma ray photons or high-energy X-ray photons are used, with an
energy range of 100-300 keV [30]. Here, one photon is detected at a time by
a single or a set of collimated radiation detectors. Today, most sensors are
based on single or multiple NaI(TI) scintillation detectors.
SPECT imaging is cheaper than PET but it is less sensitive [31].
Positron emission tomography (PET)
In PET, the radioisotope attached to a targeting molecule undergoes positron
emission decay and two oppositely directed (180°) 511 keV photons are
emitted that can be registered by a circular scanner via coincident detection.
By tracking the photons, computer simulations reconstruct threedimensional images of the source of the photons. Radioisotopes that can be
used for PET imaging include 11C, 13N, 15O, 18F, 64Cu, 62Cu, 124I, 76Br, 82Rb,
18
89
Zr and 68Ga and 18F which is the most commonly used isotope [32-34]. 18Flabeled fluorodeoxyglucose (18F-FDG) is used to assess the metabolic activity of tumors.
PET imaging has many advantages compared with SPECT, in particular
its high sensitivity and spatial resolution. The disadvantages of PET are the
relatively high costs, limited use for dual isotope imaging and that the majority of the compatible radionuclides must be produced with a cyclotron,
preferably on site because of the short half-life of the radionuclides [29, 35].
Radio-immunotherapy
Targeting cancer with radiolabeled molecules for therapy purposes has not
achieved the same success as radionuclide targeting as a diagnostic tool.
However, treatment of lymphoma patients with CD20 targeting antibodies
(e.g., 131I-Tositumomab and 90Y-Ibritumomab tiuxetan) has realized great
clinical success [36]. The clinical outcomes of radio-immunotherapy of solid
disease have been moderate but are currently undergoing broad preclinical
and clinical investigations [37-40].
The radiation used for therapy can be of alpha (e.g., 211At, 225Ac, 212/213Bi,
227
Th) or beta type (e.g., 90Y, 67Cu, 131I, 186/188Re, 177Lu), or it can be applied
as Auger electrons (e.g., 125I, 111In), resulting in cytotoxic, genotoxic and
apoptotic effects [41]. The range of negatively charged beta emitters is in the
order of a few millimeters in tissue, which is suitable for larger tumors and
metastases. Higher radionuclide concentrations of beta emitters compared
with alpha emitters are required for comparable cell killing. However, crossfire of the associated penetrating radiation along the beta particle path length
decreases the need to target each cancer cell with a radionuclide emitter.
Targeting of the nucleus is crucial for effective tumor cell killing with Auger
emitters because of the nanometer range of the tracks. By using short-range
high-LET radiation, the effect on the closely situated target cell is maximized in comparison to the distal healthy tissue. Factors that influence the
clinical success of radio-immunotherapy are the amount and quality of the
delivered radioactivity, biological and radionuclide half-life and the oxygenation state of the tumor cells by simultaneous competition against enzymatic
DNA repair mechanisms [42].
Theranostics
The youngest field of nuclear medicine is the field of theranostics, which
combines the modalities of diagnostic imaging and therapy. Theranostics
deliver therapeutic drugs and imaging vectors concomitantly within the same
dose [43]. Therefore, diagnostic imaging including response monitoring of
the disease can be followed by personalized treatment utilizing the same
19
agent. Therapeutic radionuclides that can be used for molecular imaging,
e.g., 177Lu or 90Y are of particular interest in this approach [43, 44].
Targets and targeting agents
Targets of the present study
CD44 and its variant CD44v6
The CD44 cell-surface glycoprotein plays a role in the facilitation of cellcell and cell-matrix interactions through its affinity for hyaluronic acid. In
addition, it is known to impart adhesion and is also involved in the assembly
of growth factors on the cell surface, for example, EGFR and HER4 [45,
46]. Dysfunction and/or altered expression of the protein causes various
pathogenic phenotypes. Additionally, CD44 has been shown to be highly
expressed in cancer cell subpopulations with CSC-like properties, e.g., in
HNSCC and colorectal tumors [47-50].
Figure 3. A) Gene map of human CD44. CD44s does not contain variable exons.
The exons v2-v10 are alternatively spliced. B) Simplified overview of CD44. CD44
is a transmembrane molecule, which consists of a cytoplasmic domain, and extracellular hyaluronan binding domain and variable domain.
CD44 is encoded by a single gene consisting of 20 exons. The standard form
(referred to as CD44s) is encoded by exons 1-5 and 15-20. The exons that
are lacking in CD44s are called CD44 exon isoform variants (referred to as
CD44v1-10) [51]. While evidence of exon v1 expression exist in many spe-
20
cies, in humans the exon contains a stop codon and has not been found expressed, see also Figure 3.
Additionally, 19 different splice variants have been found that are generated by alternative splicing of the CD44 mRNA, all of which are expressed
at various levels in different tissues, and the roles of these variants are not
fully understood [52]. Certain CD44 splice variants, in particular those containing CD44 exon variant 6 (CD44v6), have been associated with disease
progression, tumor cell invasion and metastasis. CD44v6 expression has
been found in several cancers, including breast, gastrointestinal, colorectal
and HNSCC [46, 51, 53, 54]. The large selection of CD44 variants combined with different expression patterns in different tissues, normal cells and
cancer types, may allow for precise tumor targeting. Several CD44v6 binders are described in the section CD44v6 targeting agents.
EGFR
The 170 kDa (mass of the monomer) large transmembrane glycoprotein
EGFR (also called ErbB1 or HER1) is a member of the HER/ErbB family,
which in addition to EGFR includes three members: HER2/ErbB2/neu,
HER3/ErbB3 and HER4/ErbB4. EGFR has several known natural ligands,
such as the epidermal growth factor (EGF), transforming growth factor alpha
(TGFα), amphiregulin, betacellulin, heparin-binding EGF-like growth factor
(HB-EGF) and neuregulins (NRG1, NRG2, NRG3 and NRG4) [17, 55].
EGFR is a receptor tyrosine kinase (RTK) connecting the extracellular space
and intracellular signal transduction. After ligand binding to the extracellular
domain, the receptor undergoes a shift from the inactive monomeric to the
active hetero- or homodimeric form followed by subsequent autophosphorylation and induction of several intracellular pathways. PI3K/AKT,
JAK/STAT, SRC/Focal adhesion kinase (FAK) and MAPK/ERK are signal
cascade pathways that can be activated through EGFR, regulating processes
in the nucleus, which lead to proliferation, inhibition of apoptosis, angiogenesis, migration, differentiation, adhesion or metastatic invasion [14, 56].
Important players in the EGFR mediated signal transduction are presented in
Figure 4A.
EGFR has been found to be up-regulated and/or modified in multiple
cancer types, promoting carcinogenesis and disease progression. Additionally, the expression of EGFR has been associated with resistance to radiation
and standard chemotherapy [30, 56], which is why it has become an important target in cancer therapy and is of great interest in cancer research.
EGFR gene expression has been shown in 25-77 % of colorectal neoplasms
and 80-100 % of HNSCCs, indicating that many patients could benefit from
EGFR targeting pharmaceuticals [57].
Two approaches for EGFR targeting are used today; the intracellular suppression of the tyrosine kinase activity, and blocking of the extracellular
21
domain by specific antibodies or antibody-like structures [58]. Several
EGFR targeting molecules are described in the section EGFR targeting
agents.
HSP90
In recent years, there has been rapid progress in the identification of new
molecular targets that could be useful for cancer therapies. Some of these
promising targets are members of the heat shock protein family, which is a
group of proteins that are induced in response to cellular stress. Their primary function is to establish protein-protein interactions, stabilize threedimensional protein structures and assist newly synthesized proteins to fold
into their correct confirmation. HSP90 is one member of the very diverse
HSP group and is named based on its size [59, 60]. Several isoforms of
HSP90 have been found in the cytoplasm and in the nucleus, including
HSP90alpha and HSP90beta. Although HSP90alpha and HSP90beta show
85 % sequence identity, HSP90beta is constitutively expressed and often
referred to as the major form. Both isoforms can be overexpressed in malignant disease [61]. HSP90 contains three functional domains, the ATPbinding, the protein-binding and the dimerizing domain. It uses a complex
chaperone regulation cycle by binding and hydrolysis of ATP and cochaperones, e.g. HSP70, p23, HSP-organizing protein (HOP) and CDC37
[62]. Currently, more than 200 HSP90 client proteins have been found; they
are involved in various cellular responses and many are activated in malignancy, as summarized in Figure 4 (e.g. cell cycle progression, migration,
growth, cell signaling, activation of transcription factors) [60, 63, 64]. While
HSP90 is required in all cells, an increased expression level of HSP90 has
been found in several hematologic and solid tumors [11, 65]. Furthermore,
tumor cells are particularly sensitive to anti-HSP90 drugs because they are
‘‘oncogene addicted’’ and require especially high levels of the chaperone.
This is mainly due to the microenvironment within solid tumors, which includes chromosome and/or microsatellite instability, hypoxia, a low pH and
an insufficient nutrient supply. HSP90 overexpression in cancer cells is associated with increased tumor cell survival, an effect that is probably due to
stabilization of oncogenic cell signaling proteins, which ultimately prohibits
apoptosis. HSP90 client proteins include mutated p53, MEK, FAK, PDGFR,
VEGFR2, KIT, ATM, ATR and MET [66-68]. In addition, HSP90 stabilizes
proteins which are known to be associated with cell cycle checkpoints like
CDK2, CDK3 or CDK4, with constitutive activated signaling proteins like
AKT and ERK, or with protection against radiation-induced cell death like
HER2, EGFR, AKT and RAF-1. Another example is the stabilization of the
constitutively auto-phosphorylated EGFR variant EGFRvIII, which lacks the
extracellular ligand-binding domain [64, 69, 70]. EGFRvIII has been shown
22
to enhance tumorigenicity by increasing proliferation and decreasing apoptosis.
Targeting and inhibition of HSP90 provides the unique possibility of
overcoming mutations in multiple downstream signaling proteins, disrupting
Figure 4. A) A schematic overview of select signal transduction proteins and signaling pathways connected with HSP90 and tumorigenesis. HSP90 client proteins (red
circle) are present at several levels and key junctions of the oncogenic signaling
cascades, implying that HSP90 targeting could affect multiple pathways simultaneously and constitute an effective treatment strategy. B) Schematic illustration of
several hallmark traits of cancer which depend on HSP90 client proteins and which
are abrogated by HSP90 inhibition.
23
feedback loops and shutting down several pathways simultaneously. The
downregulation of DNA repair proteins could lead to the sensitization of
tumor cells to radiation and ultimately be utilized for combination treatment
with radiotherapy or radio-immunotherapy, an approach that was investigated in Paper IV.
Figure 5. Schematic overview of the development process of targeting agents.
Development of radiolabeled targeting agents
The development path of new cancer drugs and targeting agents is long and
winding, and each phase involves an increase in costs and efforts [71]. Generally, the first phase consists of basic medical science studies of patient
material, leading to the identification of a pathology or biochemical process
of interest. In the second phase, a specific molecular target is sought via
molecular biology and biochemistry, which can either visualize or affect the
Figure 6. Selection of targeting agents: schematic structure and properties.
24
pathology or the process. This phase is followed by the selection of a specific targeting agent and payload (signaling or therapeutic, as described previously), which involves labeling chemistry and synthesis. After this process
has led to a viable targeting compound, the following phase consists of in
vitro and in vivo studies of, e.g., the specificity, selectivity and toxicity of
the compound. Based on these tests, either the targeting compound or labeling chemistry must be re-visited to optimize the parameters. The final phases
move from the biomedical into the clinical domain, where the compound is
evaluated in terms of toxicity (Phase 0 and I clinical trials) compared with
existing drugs/targeting compounds (Phase II, III clinical trials) [29, 72].
This process is summarized in Figure 5. The majority of the work presented
in this thesis focuses on processes 2-5 (discovery, definition and preclinical
validation).
Targeting agents
The most studied molecules for imaging and radio-immunotherapy are full
size antibodies (∼150 kDa). The use of antibodies as radio-immunotargeting
agents has many advantages. They have a fairly high affinity and are easily
and economically produced. Initial problems of, e.g., murine monoclonal
antibodies causing severe immune reactions have been eliminated by routine
humanization, or through the de novo production of human antibodies via
display methodology (e.g., phage, yeast) [34, 73]. Large targeting vectors,
like antibodies, possess desirable pharmacokinetics for radioimmunotherapy due to their slow clearance from the bloodstream and long
duration in the circulation during the targeting phase of the tumor, properties
that may cause problems for radio-immunodiagnostics for which small molecules are preferable. The structures and characteristics of several targeting
agents are displayed in Figure 6.
To further reduce immunoreactivity, Fc interactions with complement and
Fc receptors on immune system cells have been removed by depletion of the
Fc region, either through the generation of conventional enzymatically derived Fab (∼50 kDa) or F(ab’)2 fragments (∼100 kDa) or by producing recombinant fragments that lack the immunoglobulin Fc or CH2 domains [34,
74]. These fragments retain the specificity and affinity of the parental antibody. Furthermore, agents with molecular weights of less than 60 kDa are
secreted via the kidneys and provide rapid kinetics and circulating half-lives
that are measured in hours rather than days. Single-chain antibody variable
fragments (scFvs) are even smaller (∼30 kDa) than Fab fragments and provide fast clearance and tissue penetration. Depending on the application, the
monovalency of scFvs can be a substantial disadvantage. Consequently,
scFvs are coupled to generate bivalent single-chain formats, including scFvFc fusion proteins (105-110 kDa), minibodies (∼80 kDa), and diabodies
25
(∼55 kDa) [73, 75, 76]. As an alternative to the engineered antibody molecules, recombinant DNA technology enables the development of new binding proteins and the creation of large libraries of potential binders. One example of a new class of binders is the group of scaffold proteins, which includes the designed ankyrin repeat proteins (DARPins), Fynomers, Affibody
molecules and Knottins [77]. A major advantage of these molecules is, despite very high affinities, their robustness and tolerance to harsh labeling
conditions (e.g., pH, high temperature). Another class of targeting molecules
is the natural peptide ligands (5-20 amino acids) and their analogues, which
are almost exclusively used for molecular imaging. Peptides can provide
excellent high-contrast images, yet not all target receptors (e.g., HER2) possess natural ligands. Moreover, peptides can trigger antagonistic effects [77].
CD44v6 and EGFR targeting agents
CD44v6 targeting agents
Several CD44v6-targeting conjugates have been investigated preclinically
and clinically for therapeutic and imaging applications, mainly in HNSCC
[51]. These compounds displayed selective accumulation in the tumor tissue
and only minimal uptake in normal tissues like squamous epithelia, oral
mucosa, lung, spleen, kidney, bone marrow and the scrotal area [78]. The
most frequently studied anti-CD44v6 molecules are variants of the mAbs
BIWA and U36 [79-81]. However, in clinical trials of bivatuzumab (BIWA
4) attached to a highly toxic anti-microtubule agent (mertansine), the agent
bound to skin keratinocytes and resulted in severe skin toxicity with a fatal
outcome in one patient. After this Phase I clinical trial, all development of
bivatuzumab mertansine was stopped [82, 83]. In its place, radioimmunotargeting of CD44v6 appeared to be a more promising strategy because radionuclides with the appropriate half-life and energy can avoid skin
toxicity [84]. A radio-immunotherapy Phase I trial with the 186Re-labeled
chimeric mAb (cmAb), cU36, resulted in promising anti-tumor effects and
stabilized the disease without skin toxicity [85]. PET studies in HNSCC
patients with 89Zr-cU36 permitted the visualization of primary disease as
well as metastases in the neck region and was well tolerated by all of the
subjects [86].
In addition, the use of radiolabeled antibody fragments, e.g., F(ab’)2 and
Fab’ fragments, was even superior to the parental U36 mAb in terms of tissue penetration and tumor to blood ratios at early time points [87]. The current in vitro design and selection based on recombinant combinatorial libraries is a promising strategy for the development of new CD44v6 binders with
novel, superior characteristics [88].
26
EGFR targeting agents
The EGFR serves as an attractive target for molecular imaging and therapy,
mainly due to the strong correlation between its deregulation and the aggressiveness of the disease [89]. Numerous EGFR targeting compounds are approved by the FDA or in ongoing clinical trials. These drugs either target the
extracellular domain of the cell surface receptor or the intracellular tyrosine
kinase domain, the so-called tyrosine kinase inhibitors (TKIs). Both the targeting of the extra- and intracellular domains has been used for therapy and
molecular imaging.
The most commonly used EGFR targeting drug is cetuximab (Erbitux,
ImClone System Inc.), a chimeric IgG1 mAb that blocks receptor activation
and downstream signaling by binding to the extracellular ligand-binding
domain and inducing internalization [32, 89]. Cetuximab is used as single
agent or in combination with radiotherapy in several solid cancers, e.g.,
HNSCC and colorectal cancer. Still, the therapeutic potential of the targeting
of EGFR remains to be refined and optimized [57]. Two major approaches
have been undertaken for the development of EGFR tracers for molecular
imaging: labeling of mAbs and their derivatives and small organic compounds.
Cetuximab labeled with various radionuclides has been used in SPECT
and PET cameras [90]. 89Zr-cetuximab and 177Lu-cetuximab investigated by
PET before radio-immunotherapy confirmed the tumor targeting and allowed the estimation of radiation dose delivery to tumors and normal tissues
[91]. The expression and quantification of EGFR has been investigated with
86
Y-labeled and 64Cu-labeled cetuximab [90, 92]. Furthermore, 111Incetuximab-F(ab’)2 imaging allowed the effects of EGFR inhibition to be
monitored in combination with radiation treatment in HNSCC models [93].
Another approach to anti-EGFR antibodies and their derivatives is imaging
with natural ligands such as human epidermal growth factor (hEGF, approximately 6.4 kDa) [94]. Also EGF has been labeled with various SPECT radionuclides, including 99mTc [95] and 111In [96]. Small animal PET scans with
68
Ga-labeled DOTA-hEGF or 18F-FBEM-EGF could visualize EGFR expressing tumors within 5 min to 2 h p.i. [97, 98]. Furthermore, radiolabeled
small scaffold molecules [77] or tyrosine kinase inhibitors are interesting
anti-EGFR targeting vectors. For example, several Affibody molecules targeting EGFR have demonstrated preferable biodistribution patterns despite a
high kidney uptake, as well as superior imaging properties [99, 100]. Moreover, targeting of mutated EGFR variants with different targeting vectors,
especially EGFRvIII, is receiving increasing interest in nuclear medicine
[73, 101].
27
HSP90 inhibitors
At present, HSP90 inhibitors are gaining increasing preclinical and clinical
attention due to their unique potential for combined targeting of multiple
oncogenic protein pathways. Since the initial discovery of the first natural
product HSP90 inhibitors, including geldanamycin and radicicol, almost two
decades ago, remarkable progress has been achieved in the development of
effective and selective drugs against this chaperone [102, 103]. The mode of
action for all developed anti-HSP90 compounds is very similar. They bind to
the regulatory ATP/ADP pocket in the amino-terminal portion of HSP90,
blocking the unfolded client protein from binding and thus causing its ubiquitination and proteasomal degradation, which may lead to apoptosis. Additionally, HSP90 inhibition can trigger the recognition and destruction of
cancer cells by the immune system of the patient because the accumulation
of misfolded proteins leads to digestion into small peptides, which are presented on the cell surface by MHC class I molecules [104].
It has been found that instead of complex molecules such as radicicol,
smaller molecules are more efficient [105]. The synthetic geldanamycin
analogs, e.g., 17-Allyl-17-Demethoxygeldanamycin (17-AAG), 17(dimethylaminoethylamino)-17-demethoxygeldanamycin
(17-DMAG),
NVP-AUY922 and NVP-BEP800, have been intensely characterized in vitro
and in vivo [106-109]. However, the clinical outcomes of the previously
described drugs have been modest due to poor pharmaceutical properties,
limited solubility in water, difficulties associated with their formulation and
high hepatotoxicity [110]. New synthetic small molecules, second- and
third-generation HSP90 inhibitors, are currently developed via the combined
use of high-throughput screening and structure-based drug design. These
high-affinity HSP90 inhibitors may overcome the limitations of the previously described drugs because they are easier to administer and may have
reduced toxicity. One of these promising, novel and high-affinity HSP90
inhibitors is AT13387 (2,4-dihydroxy-5-isopropyl-phenyl)-[5-(4-methylpiperazin-1-ylmethyl)-1,3-dihydro-isoindol-2-yl] thanone, l-lactic acid salt)
[64, 111]. AT13387 is now undergoing evaluation in Phase I and II clinical
trials that include patients with prostate carcinomas, refractory tumors, gastrointestinal stromal tumors (GIST) and ALK positive lung cancer [112,
113]. Studies on AT13387 are presented in Paper IV and Paper V.
HSP90 inhibition in combination with other treatments
The main aims of treatment combinations are to achieve synergistic therapeutic effects that account for the potential tumor heterogeneity and provide
dose and toxicity reductions and minimize the induction of drug resistance.
The development of resistance in particular is a major problem in cancer
28
patient management and is frequently induced by rapid genetic and epigenetic changes during adaptation to the microenvironment in the tumor or induced by the treatment itself [63]. Targeting and inhibiting HSP90 represents a novel approach to overcome resistance, especially in combination
with other treatment options. Preclinical and clinical studies have demonstrated additive and synergistic effects when combining HSP90 inhibitors
with other anti-cancer drugs in solid and hematologic tumors [63, 114]. In
HER2-positive breast cancer patients who are progressing on the HER2targeting drug trastuzumab (HerceptinTM), proof-of-concept was demonstrated by first-generation HSP90 inhibitors in combination with trastuzumab
[114].
Furthermore, the inhibition of HSP90 is an interesting strategy to sensitize tumors towards irradiation. The misfolding, degradation and finally
depletion of HSP90 client proteins is associated with antitumor activity and
may also radiosensitize cells, because the cell cycle control and DNA repair
machineries, among others, are affected [115-118]. Interestingly, several
preclinical studies have demonstrated that first generation HSP90 inhibitors
do not radiosensitize normal cell lines [116, 119].
In effect, this approach could lead to lower radiation dosages or fewer radiation treatments and potentially reduce systemic exposure and undesirable
side effects in normal tissues. Furthermore, this strategy may be useful for
restoring efficacy in chemotherapy and radiotherapy-resistant cancers and/or
reducing the recurrence of disease.
Current preclinical findings, including the results in Paper IV, indicate
that HSP90 inhibitors have promising efficacy as an adjunct to radiation
therapy in several types of cancer [105, 117, 118]. Although the radiosensitizing effects of HSP90 inhibitors have been shown in vitro, this concept has
not been investigated in a clinical setting. The next logical step is to translate
these findings to Phase 0/Phase I clinical studies investigating this therapeutic combination.
29
Aim
The overall aim was to characterize the heterogeneity of tumor cells and to
evaluate new biomarkers for radio-immunodiagnostics and radioimmunotherapy. There is a need to develop new tracers that target these
biomarkers and combine treatment strategies to potentiate the synergistic
effects between new cancer drugs and radio-immunotherapy, which could
lead to improved treatment protocols.
The specific goals were as follows:
30
•
Evaluate new potential targets for radio-immunotherapy and diagnostic imaging, with a focus on the expression fingerprints of CD44
variants
•
Investigate radiolabeled CD44v6-targeting fragments as targeting
agents for radio-immunodiagnostics (PET and SPECT imaging) of
CD44v6-expressing tumors
•
Characterize the novel HSP90 inhibitor AT13387 in vitro and in vivo with regard to its potential radiosensitizing effects
•
Evaluate biomarkers for imaging and radio-immunotherapy in vivo
in combination with HSP90 inhibition
Results
Paper I
New targets for imaging and radio-immunotherapy:
CD44 variants
Aim and background
Several studies have found high expression of the molecules CD133 and
CD44 and low expression of CD24 to be markers for aggressive subpopulations in various malignancies [8].
The aim of the present study was to identify specific expression fingerprints of these markers, with a special focus on CD44 and its variants
CD44v3, v4, v5, v6, v7, v8 and v10, in squamous cell carcinoma cell lines,
to identify suitable targets for molecular radiotherapy or diagnostic imaging.
Furthermore, we wanted to determine whether the expression of these variants changes under different growth conditions, such as a lack of nutrients
(as commonly exists inside solid tumors, simulated by serum starvation),
and whether any of the variants is associated with a certain phase of the cell
cycle, increased proliferation, migration potential and radioresistance. Such
molecular fingerprints could, e.g., be used for patient stratification, therapeutic monitoring, or selective targeting of specific tumor subpopulations.
Table 2. Expression pattern of CD44, CD44 exon variants, CD133, and CD24. The
expression levels were measured by flow cytometry and graded on a relative scale to
the isotope control (expression baseline).
– low expression; + moderate expression, ++ high expression; n = 2-6.
Cell line
UT-SCC7
UT-SCC12
SCC-25
H314
Expression of the marker
CD133 CD24 CD44 v3
v4
v5
v6
v7
v7/8
v10
+
+
+
+/-
++
++
++
+
+
+
-
++
++
++
+
++
++
++
++
-
+
+
+/-
-
++
++
++
++
++
++
+
+
31
Methods
The HNSCC cell lines were characterized in vitro under standard and low
serum culturing conditions. Cell proliferation and radiation resistance were
studied using clonogenic survival assays. The migratory activity of starved
and non-starved cells was assessed using a cell migration scratch (woundhealing) assay. Morphology, antigen expression and cell cycle distribution
were assessed by flow cytometry and fluorescence-activated cell sorting
(FACS) using DNA binding dyes and fluorescently labeled antibodies.
Figure 7. A) Example of flow cytometry analysis of three markers with different
expressed levels in the HNSCC cell line SCC-25. CD24 displays no or low expression. CD44v3 is moderately expressed. CD44v6 exhibits a high expression level. B)
Clonogenic survival assay of SCC-25 cells under normal growth conditions and
after serum starvation for 4 months (1 % FBS, 20 ng/ml EGF and bFGF). Normal
SCC-25 cells are more sensitive to radiation than previously starved SCC-25 cells.
The survival data were fit to a linear quadratic curve. Error bars represent the
standard deviation (SD), n ≥9.
Results
The investigated HNSCC cell lines lacked or displayed very low expression
patterns for CD24. CD133 was moderately expressed in three of the four cell
lines, whereas all of the cell lines were highly positive for CD44 expression
(see Table 2 and Figure 7A). High and uniform expression levels of the
CD44 standard variant (CD44s) and the variants CD44v6 and CD44v7 were
measured in all of the cell lines. CD44v3 was highly expressed in some of
the studied cell lines, whereas CD44v5 and CD44v8 demonstrated low or no
expression. In several cell lines, an interesting subpopulation of very high
antigen expression was identified for the variant CD44v4, which was independent of the cell cycle phase. In the cell starvation experiments that were
designed to simulate the environment within a large tumor and to increase
the numbers of aggressive cancer cells, a major population with dramatically
increased expression of CD44, CD44v6 and CD44v7 was identified. Additionally, the morphology of the starved cells was altered, rendering fewer
cells that grew as a monolayer and a larger proportion of cells that grew
32
three-dimensionally. Previously starved CD44++, CD44v6++ and
CD44v7++ cells displayed enhanced motility and increased resistance to
radiation compared with non-treated cells (Figure 7B).
Conclusion and discussion
By analyzing novel molecular tumor biomarkers, we hope to elucidate the
molecular and biological mechanisms responsible for tumor progression and
aggressiveness and to identify suitable tumor-exclusive targets that can distinguish aggressive, resistant, and re-growing cancer subpopulations. Successful selective targeting of these subpopulations could lead to improved
therapeutic responses and a reduced recurrence of cancer.
The characterization of CD44 and its variants revealed that CD44,
CD44v6 and CD44v7 are highly expressed in cultured HNSCC cells, in
which they were found to mediate migration, proliferation, and radiation
resistance. The markers CD44v6 and CD44v7 are very interesting because
they are more specific anti-tumor targets than the commonly assessed
CD44s, which is also present to a high extent in normal tissue. We conclude
that targeting splice variants containing v6 and/or v7 could provide a basis
for more personalized medicine in the future, hopefully enabling improved
treatment outcomes together with a better quality of life and longer lifetime
expectancies in patients with HNSCC.
33
Paper II and III
Radiolabeled CD44v6-targeting fragments for radioimmunodiagnostics
Aim and background
Paper I demonstrated that the targeting of CD44v6 could be a viable strategy
for radio-immunodiagnostics in HNSCC. For molecular imaging, small tracers such as antibody fragments directed against cell surface molecules appear to be a particularly favorable class of molecules due to the higher contrast and earlier optimal imaging time compared with the full-length parental
antibodies.
Therefore, we have developed a fully human, anti-CD44v6 Fab fragment,
AbD15179, and a bivalent antibody fragment, AbD19384, engineered from
two AbD15179 Fab-fragments corresponding in size to a F(ab’)2.
The objective of this study was to evaluate, for the first time, the in vitro
and in vivo properties of radiolabelled AbD15179 (Paper II) and of the bivalent antibody fragment AbD19384 (Paper III) as novel targeting tracers for
radio-immunodiagnostics of CD44v6-expressing tumors.
Methods
In Paper II the Fab fragment AbD15179 was labeled with 111In and 125I as
models for radionuclides that are suitable for molecular imaging with
SPECT or PET, respectively. In vitro studies resulted in the characterization
of AbD15179 in terms of the species and antigen specificity as well as the
internalization properties. The in vivo specificity and biodistribution were
then evaluated in a mouse xenograft model using a dual-tumor and dualisotope approach.
In Paper III, the specificity, binding properties, and interaction analysis of
the bivalent antibody fragment AbD19384 were assessed in vitro. 125Ilabeled AbD19384 was then studied in mouse xenografts bearing two tumors with different expression levels of CD44v6. Small animal PET/CT
scans revealed the uptake and distribution of 124I-labeled AbD19384 and 18FFDG in mice bearing tumors with both low and high CD44v6 expression at
three different time points.
Results
AbD15179 was successfully labeled with both 111In and 125I using the chelator CHX-A″-DTPA [120, 121] and a direct labeling approach with chloramine T (CAT) [88], respectively. Both the radiolabeled AbD15179 conjugates 111In-Fab and 125I-Fab demonstrated selective tumor cell binding in
34
Figure 8. A) Biodistribution of 111In-Fab and 125I-Fab as percentages of injected
activity per gram of tissue (%ID/g), excluding the thyroid (inset), which is expressed
as percentages of injected activity per organ. B) Tumor-to-organ ratios for A431
tumors and inset: tumor to blood ratios of radiolabelled Fab for both A431 and
H314 tumors. The target antigen CD44v6 was expressed moderately in H314 tumors and at high levels in A431 tumors. The animals were sacrificed at 6 h (black
bars), 24 h (dark grey), 48 h (light grey) and 72 h (white) post injection. A431 tumors were used as reference for tumor to organ ratio calculations. The animals
were sacrificed at 6 h (black bars), 24 h (dark grey), 48 h (light grey) and 72 h
(white) post injection. Error bars represent SD, n = 5.
35
vitro and good tumor targeting properties in vivo (Figure 8A). However,
111
In-Fab displayed higher tumor uptake, slower dissociation as well as
higher tumor to blood ratio compared to 125I-Fab (Figure 8B). On the other
hand, uptake in non-target organs like liver, spleen and kidneys was lower
for iodinated Fab.
Figure 9. Representative data from SPR analyses of A) AbD15179 and B)
AbD19384 binding to CD44v3–10. Five concentrations of each fragment ranging
from 3 to 50 nM were injected over a surface with immobilized CD44v3–10 on a
ProteOn XPR36 protein interaction array system Bio-Rad. Data are double referenced by subtraction of simultaneous responses from a blank surface and a buffer
injection. Experimental data are plotted together with curves drawn from a fitted
1:1 Langmuir isotherm.
AbD19384 was successfully conjugated with either 125I (using direct chloramine T labeling) or with 124I (using iodogen labeling (1,3,4,6-Tetrachloro3α,6α-diphenylglycouril)). SPR analysis confirmed specific binding of
AbD15179 and of AbD19384, where AbD198384 displayed a higher affinity
combined with improved dissociation rate compared to AbD15179, see Figure 9. Ligand Tracer measurements showed that 125I-labeled AbD19384 was
able to distinguish between CD44v6 high, moderate and low expressing
cancer cells (Figure 10A). The labeling method (CAT or iodogen) did not
influence the binding properties of the tracer (Figure 10B).
Figure 10. Representative Ligand Tracer measurements of real-time binding of
radioiodinated AbD19384. Binding traces using three subsequent concentrations
(10, 30, 90 nM) were obtained for at least 1 h per concentration (marked by dotted
lines), followed by a dissociation measurement for at least 15 h. A) Binding of 125IAbD19384 to A431, H314, UM-SCC-74B, or negative MDA-MB-231 cells. Signal is
shown in cps. B) Comparison of 125I-AbD19384 (labeled using CAT), 125I-AbD19384
(labeled using iodogen) and 124I-AbD19384 (labeled using iodogen). Curves were
normalized to the percentage of maximum binding.
36
Furthermore, 125I-AbD19384 displayed a favorable biodistribution and tumor-specific uptake, which was consistent with the small animal PET/CT
study investigating 124I-labeled AbD19384, as summarized in Table 3. 124IAbD19384 clearly allowed the visualization of high CD44v6-expressing
tumors, while 18F-FDG was neither able to detect nor distinguish between
CD44v6 high and low-expressing tumors. The contrast and tumor to blood
ratios increased with time (up to 72 h) though 48 h p.i. was optimal for imaging (Figure 11).
Conclusion and discussion
We conclude that CD44v6 expression profiling has the potential to be a valuable diagnostic tool in HNSCC patients. We could prove, for the first time,
that the novel radiolabeled fragment AbD15179 was able to efficiently target
CD44v6-positive tumors in vitro and in an in vivo setting. In particular,
111
In-Fab showed favorable tumor to blood ratios. Furthermore, the results of
Paper II demonstrate that radiolabeling can change the kinetic properties of
the tracer, emphasizing the importance of repeated characterization of the
protein even after radiolabeling.
Table 3. Comparison of tumor uptake of 124I-AbD19384 relative to blood (heart), as
obtained by PET imaging and ex vivo organ distribution. 24 h, 48 h and 72 h refer to
time points post administration of 124I-AbD19384.
Tumor type
A431
UM-SCC-74B
Mode
PET
Ex vivo
PET
Ex vivo
24 h
Mean SD
N
Mean
48 h
SD
N
72 h
Mean SD
N
0.83
0.71
0.33
0.34
4
4
3
4
1.32
1.86
0.49
0.52
0.32
0.71
0.36
0.38
3
4
3
4
1.44
1.83
0.99
0.90
3
4
3
4
0.18
0.17
0.03
0.04
0.58
0.64
0.29
0.29
The bivalent antibody fragment AbD19384 was also found to be a promising
candidate for the imaging of high CD44v6-expressing tumors. Functional
anti-CD44v6 imaging with AbD19384 revealed many advantages compared
with imaging using the clinical standard 18F-FDG, although since only one
animal was used, the FDG scan should only be viewed as descriptive.
Based on these results, radiolabeled CD44v6 targeting fragments could be
used in the future for patient stratification and the early detection of HNSCC
in the clinic. Therefore, they could help to improve the management of head
and neck malignancies.
37
Figure 11. Small animal PET/CT imaging of 124I-AbD19384 in CD44v6 high expressing tumors on the left (T1) and low expressing tumors (T2) on the right flank.
The best contrast was obtained at 48 h p.i.. 18FDG-PET shown on the right at 30
min p.i.. 18F-FDG demonstrates no clear visualization of tumors, no discrepancy
between the high CD44v6 expressing tumor and the low expressing tumor, and
uptake in brown fat (B).
38
Paper IV
The HSP90 inhibitor AT13387 potentiates the effects of
radiation
Aim and background
HSP90 overexpression has been demonstrated in HNSCC and adenocarcinomas of the colon. Overexpression is associated with increased tumor cell
survival due to the stabilization of oncogenic client proteins. Inhibition of
HSP90 provides the possibility to target multiple oncoproteins concurrently
and to disrupt several cell-signaling pathways, including feedback loops.
The aim of this study was to assess the treatment efficacy of the novel
HSP90 inhibitor AT13387, alone or in combination with radiotherapy. A
special focus was to investigate the effects of this inhibitor on migration, the
cell cycle and the expression of oncogenic client proteins in vitro in SCC
and colorectal cancer cells, with the goal of exploring the mechanisms underlying radiosensitization. Furthermore, we aimed to investigate the in vivo
treatment response of commonly expressed cell surface receptors, cell signaling and DNA repair proteins in mouse xenografts.
Methods
Colony-forming and multicellular tumor spheroid assays, flow cytometry,
scratch (wound-healing) assays, Western blot analysis of target antigen expression and Pulsed-field gel electrophoresis (PFGE) were first used to evaluate the effect of AT13387 on cell survival, motility, cell cycle distribution,
radiosensitivity and DNA repair capabilities in vitro. Potential radiosensitizing effects of the inhibitor were studied in 2D and 3D cell cultures after exposure to external beam radiation. Effects on antigen expression were then
assessed in vivo in mouse xenografts bearing CD44v6 high and EGFR high
tumors. The animals were treated 5 times on 5 consecutive days with 50
mg/kg AT13387. The tumors were dissected and analyzed using immunohistochemistry (IHC).
39
Figure 12. Clonogenic survival and Multicellular tumor spheroid growth. A) Clonogenic survival assays. Dose response curves of H314, A431 LS174T and HCT116
cells treated with AT13387 (0.5 nM, 5 nM, 50 nM) and radiation (2, 4 and 6 Gy).
The cells were pre-plated in triplicates, incubated with AT13387 24 h later and
irradiated 1 h after drug incubation. Colonies with >50 cells were counted. (n ≥ 612). All curves are normalized to the plating efficiency of the non‐irradiated controls. B) Effects of AT13387 and radiation alone as measured by plating efficiencies
of the dataset in A), evaluated with Student’s t-test with * p < 0.05, ** p < 0.01, ***
p < 0.001 C) Multicellular tumor spheroid growth. H314 cells and HCT116 cells
treated with AT13387 (5 nM, 50 nM, 100 nM), 5 times 2 Gy radiation fractions and
combination treatment of 5 nM AT13387 and radiation. 1000-3000 cells were preplated in agarose coated 96-well plates, incubated with AT13387 after 24 h and
irradiated 1 h after drug incubation. The error bars represent SD, n ≥ 3. All curves
are normalized to the size of controls at day 1. D) Example of H341 spheroids after
2, 12 and 22 days.
40
Figure 13. DNA DSB rejoining capacity, migration distance and cell cycle analysis.
A) PFGE analysis. H314, A431, LS174T and HCT116 cells were exposed to 200 nM
AT13387 for 24 h prior 40 Gy radiation. After irradiation, cells were allowed to
repair. Kinetics of DSB end rejoining was calculated by fraction of activity released
(FAR) corresponding to DNA of sizes < 5.7 Mbp. The error bars represent SD, n =
4. B) Cell migration assay. Left hand images represent photographs of A431 and
HCT116 cultures taken at 0 h (immediately after scratching) and at 24 h with and
without AT13387 treatment. The graphs show quantification of the wounded area
invaded after 24 h, measured in migrated distance in mm. The error bars represent
SD, n ≥ 3. Student's t-test was used to calculate statistics: * p < 0.05, ** p < 0.01,
*** p < 0.001. C) Flow cytometry evaluation of cell cycle progression. The histograms on the left show representative data of DNA content stained with DAPI/PI to
show the progression from G1 through the S and G2/M phases. Increasing concentrations of AT13387 resulted in a greater G2/M peak and depletion of the S phase.
The panel on the right shows the quantification of the flow cytometry data and statistical significance based on ANOVA with Tukey’s post-test.
41
Results
AT13387 treatment displayed effective cytotoxic activity in colorectal and
SCC cells with low nanomolar IC50 values. AT13387 significantly radiosensitized the tested cells in 2D and 3D cell culture models, as seen in Figure
12. HSP90 client proteins were efficiently downregulated, both in vitro and
in vivo. Moreover, exposure to AT13387 resulted in G2/M phase arrest,
depletion of S phase and significantly reduced migration ability (Figure 13).
Immunohistochemical staining of tumor tissue is presented in Figure 14.
AT13387 treatment efficiently downregulated HSP90, tumor markers like
EGFR and MET, and DNA repair proteins like DNA-PKcs and ATM.
CD44v6 expression was not significantly altered by the drug, however.
Figure 14. Immunohistochemical analysis of A431 tumors. Representative images of
ex vivo immunohistochemical stainings for HSP90, EGFR, CD44, CD44v6, DNA‐
PKcs, ATM and MET expression on A431 tumor xenografts (magnification x10).
Mice in the treatment group (n = 6) received 5 doses of 50 mg/kg AT13387 on 5
consecutive days before dissection and analysis. The effect of AT13387 was highest
on the expression pattern of HSP90, EGFR and MET.
Conclusion and discussion
We conclude that AT13387 is a potent new cancer drug with improved
pharmacokinetics compared with first-generation HSP90 inhibitors. We
showed for the first time the excellent anti-tumor effects of AT13387 alone
and in combination with radiation in SCC and in colorectal cancer cells both
42
in vitro and in vivo. The synergistic combination effects at clinically relevant
drug and radiation doses are especially promising for both reduction of the
radiation dose and minimization of side effects, or for an improved therapeutic response.
The mechanism underlying this effect is likely related to the detected
downregulation of HSP90 client proteins, which are involved in all hallmarks of cancer. Our results strengthen the case for further clinical studies of
HSP90 inhibitors and radiation co-treatment.
Furthermore, the varying treatment responses of cell surface proteins
could potentially be used either as biomarkers for the monitoring of
AT13387 treatment or as potential targets for radio-immunotherapy when
unaffected by HSP90 inhibition.
43
Paper V
Molecular imaging in combination with HSP90
inhibition
Aim and background
Paper IV demonstrated that the novel HSP90 inhibitor AT13387 promotes
the degradation of oncogenic proteins upon binding and acts as a radiosensitizer. However, for optimal treatment, there is a need to identify biomarkers
for therapeutic response monitoring, patient stratification and to find suitable
targets for combination treatments.
Molecular imaging of shifts in biomarker expression after AT13387
treatment could be a suitable approach to assess the treatment response. One
marker for treatment response monitoring identified in Paper IV is of specific interest: EGFR. EGFR expression has been associated with tumor progression and the emergence of chemo- and radiotherapy resistance in several
cancer types.
Furthermore, combined treatment modalities such as AT13387 together
with radio-immunotherapy might potentiate treatment outcomes due to the
radiosensitizing effects of the drug. Paper I showed that CD44v6 is an interesting overexpressed biomarker in HNSCC, and Paper IV demonstrated that
CD44v6 was not affected by HSP90 inhibition, implicating CD44v6 as a
potential marker for targeted radionuclide therapy in combination with
AT13387 in HNSCC.
The aim of this study was to target and monitor EGFR and CD44v6 with
suitable radiotracers using PET/CT in animals treated with AT13387 to establish whether they are viable biomarkers for this purpose. The expression
of EGFR and CD44v6 was investigated using 124I-labeled Cetuximab and
124
I-AbD19384, respectively. 124I-AbD19384 is a novel anti-CD44v6 tracer
that was initially investigated (Paper III) in mouse xenografts with
EGFR/CD44v6 high and low-expressing SCC tumors. For comparison of the
tracers and of the specificity, the metabolic activity of the tumors in control
and treated mice was concurrently investigated using the clinical standard
18
F-FDG.
Methods
Cancer cell proliferation, cell toxicity (MTT) assays and radioimmunoassays with 125I-cetuximab and 125I-AbD19384 were used to quantify
the effect of AT13387 on EGFR and CD44v6 expression in vitro. Inhibitor
effects were then assessed in vivo in mice xenograft bearing tumors with
high EGFR/CD44v6 and low EGFR/CD44v6 expression. Animals were
treated 5 times on 5 consecutive days with AT13387 (50 mg/kg), and were
44
then imaged either with 18F-FDG (30 min p.i.) or with 124I-labeled tracer (48
h p.i.), using small animal PET/CT, followed by ex vivo biodistribution
measurements and immunohistochemical analysis.
Figure 15. Expression of EGFR and CD44v6 in A) A431 and B) UM-SCC-74B cells
using radio-immunoanalysis. Cells were exposed to 0.01 to 60 nM of 124I-cetuximab
or 124I-AbD19384 and a 100-fold excess of unlabeled antibody was added at the
highest concentrations for unspecific binding correction. The number of cells was
counted and radioactivity measurements were performed in a gamma counter, n =
3, error bars represent standard error of the mean (SEM).
Results
Cancer cell treatment with AT13387 caused sufficient cytotoxicity and radiosensitization with low IC50 values, below 4 nM. In vitro measurements
showed specificity of the radioiodine labeled compounds cetuximab and
AbD19384 and downregulation of EGFR after AT13387 exposure in high
and low expressing cancer cells, while CD44v6 expression was not affected
(Figure 15). Quantitative PET analysis of EGFR with the 124I-labeled mAb
cetuximab demonstrated a significant reduction of EGFR in the AT13387
treated group compared to untreated mice (Figure 16A). CD44v6 expression
(visualized with 124I-labeled AbD19384) and 18F-FDG uptake were not significantly altered by AT13387 treatment (Figure 16B and C).
45
Figure 16. Representative small animal PET/CT images of EGFR and CD44v6 high
expressing A431 tumors (T1 left posterior flank) and EGFR and CD44v6 low expressing UM-SCC-74B tumors (T2 right posterior flank) in nude mice after intravenous injection (i.v.) of A) 124I-cetuximab, B) 124I-AbD19384, C) 18F-FDG. The upper
row shows a representative cross section of the xenograft. The tracer uptake was
calculated as quotient of high and low expressing tumors. The lower row displays
planar maximum intensity projections (MIP) images of the tracer distribution.
Conclusion and discussion
In conclusion, CD44v6 is not dependent on the molecular chaperone HSP90,
and therefore, radio-immunotargeting of CD44v6 in combination with the
HSP90 inhibitor AT13387 might potentiate treatment outcomes due to the
radiosensitizing effects of the drug. A combination approach may allow a
decrease in the radiation doses to normal and dose-limited tissues and may
overcome resistance to conventional chemotherapeutic and radiation therapies.
In contrast to CD44v6, EGFR expression levels correlate with HSP90 inhibition, and molecular imaging of EGFR-positive SCC may be used to assess treatment responses to HSP90 inhibitors. Our results indicate that molecular imaging may serve as a tool to monitor responses and complement or
replace standard tumor size measurements. Currently, treatment outcomes
are measured according to RECIST criteria (response evaluation criteria in
solid tumors), which relies to a great extent on the size of the tumor. This
could be misleading in many ways, e.g., when the main bulk of the tumor
consists of non-tumorigenic cells that are more easily killed. PET imaging
for the treatment response has many advantages because it is noninvasive,
repeatable and permits the investigation of the whole tumor burden in the
body. To further investigate the AT13387 treatment response, repeated PET
analysis could be beneficial because the drug-induced effects on biomarkers
are temporary, as shown in Paper IV. We believe that treatment follow-up
with suitable PET tracers, such as mAb Cetuximab or even smaller conjugates, is likely to be an important method for estimating the duration of the
treatment effect as well as for defining personalized drug doses and sched46
ules. Furthermore, imaging of response biomarkers has the potential to serve
as an early indicator of treatment adaptation and the development of resistance.
47
48
Concluding remarks
This thesis has focused on new diagnostic biomarkers and on radiosensitization strategies following HSP90 inhibition as a step towards more personalized cancer medicine.
A specific focus has been the screening for novel molecular targets such
as CD44 exon variants, which were found to provide unique expression patterns in squamous cell carcinoma cells. Furthermore, several new radioconjugates that were tailored to target CD44v6 were evaluated in vitro and in
vivo in terms of their specificity, biodistribution and imaging performance.
The final part of the work presented herein concerns the new targeted
chemotherapeutic compound AT13387 and the potential to use it in combination with radiation treatment. The AT13387 investigation provided promising targets for treatment response monitoring and targeted radionuclide
therapy.
The main findings of the thesis are as follows:
•
•
•
•
•
•
•
High and homogenous expression of the cell surface protein CD44 and
its variants CD44v6 and CD44v7 in HNSCC cell lines
Serum starvation increases the amount of aggressive cancer cells, which
also displays an increased expression of CD44, CD44v6 and CD44v7
Cells enriched under starvation show increased migration and higher
radioresistance in comparison to non-starved cells
The novel Fab fragment AbD15179 is specific for CD44v6 with high
affinity and retention
AbD15179 can be labeled with both 111In and 125I using the chelator
CHX-A″-DTPA or a direct labeling approach with chloramine T, respectively with sufficient yield and serum stability and no detectable internalization
The labeling chemistry affects performance parameters such as tumor
uptake, dissociation rate and tumor to organ ratios. Indium labeling of
AbD15179 induces higher uptake with high tumor to blood ratio compared to iodine labeling, which in turn has less uptake in non-target organs like liver, spleen and kidneys.
The bivalent fragment AbD19384 is specific for CD44v6 with higher
affinity compared to AbD15179 and pronounced retention in vitro
49
•
•
•
•
•
•
•
•
•
•
•
•
•
50
AbD19384 can be successfully labeled with both 125I and 124I using direct labeling with chloramine T and iodogen with sufficient yield and serum stability
125
I-AbD19384 shows favorable biodistribution with a similar tumor and
organ uptake pattern as the previously described Fab fragment 125IAbD15179
124
I-AbD19384 is a promising PET tracer showing best imaging qualities
after 48 h
124
I-AbD19384 is able to discriminate between CD44v6 high and lowexpressing tumors, which cannot be achieved with the current imaging
standard 18F-FDG
The HSP90 inhibitor AT13387 is a potent new cancer drug with highly
cytotoxic effects on SCC and colon cancer cells
Co-treatment of radiation and AT13387 triggers synergistic effects,
leading to increased efficacy and tumor cell kill
AT13387 decreases cell mobility rates and causes G2/M cell cycle arrest
and simultaneous S-phase depletion, without affecting DNA DSB rejoining capacity after radiation exposure
AT13387 efficiently downregulates HSP90 and its oncogenic client
proteins DNA-PKcs, ATM, EGFR, MET and AKT involved in DNA repair, cell signaling, growth and proliferation
The effect of AT13387 on the expression levels of client proteins is transient and recurs after 24 h in the absence of drug exposure
CD44v6 is not stabilized by the chaperone HSP90 and therefore is not
affected by AT13387 treatment
The treatment response of AT13387 can be visualized by targeting
EGFR with 124I-Cetuximab using PET/CT to quantify the expression
level, whereas the treatment effect cannot be distinguished using the current clinical standard 18F-FDG
CD44v6 is a promising target for patient stratification in HNSCC
CD44v6 is a promising target for radio-immunotherapy and AT13387
co-treatment due to the lack of downregulation after HSP90 inhibition
Future studies
We believe that future studies should be directed along several lines of research. First, the results from Paper I must be confirmed and expanded upon
using a broader variety of tumor strains and patient material. Such a study
should be centered on the large-scale expression analysis of CD44 variant
isoforms, which display fingerprint-like features that we anticipate to be
associated with distinct patient outcomes. These findings could be used for
detection or therapy targeting in a variety of cancer types.
All five studies (Papers I-V) presented in this thesis rely on the wellestablished method of cancer cell line cultivation. The use of immortalized
cancer cell lines in cancer research is a valuable tool for the investigation of
cancer, but this method has many limitations. These limitations include the
loss of heterogeneity and of genetic alterations over time with extensive in
vitro passaging. The use of primary cancer cells derived from solid clinical
specimens collected at the time of surgery could be a useful technique to
avoid such pitfalls. Such primary cells can be used in 2D and 3D cell culture
experiments as well as in mouse xenografts. Despite the representation of
the original specific clinical specimen, another advantage is that such primary cells could be sampled from different locations in the patient, e.g., the
primary tumor and a different metastasis. Therefore, the establishment of
primary tumor cell models, e.g., HNSCC patients, is an attractive approach
for future investigations.
Paper II and Paper III demonstrated that radio-immunotargeting of
CD44v6 is a promising strategy for early non-invasive diagnosis and
HNSCC patient stratification. The investigated conjugates AbD15179 and
AbD19384 displayed favorable kinetics, but their targeting properties could
be improved. It would be valuable to direct focus toward optimizing the
choice of radionuclide and the radiolabeling method. Furthermore, the flexible recombinant origin of AbD19384 might facilitate the development of
novel tracers that are adapted either for targeted therapeutic or diagnostic
applications, which may help improve the management of head and neck
malignancies in the future.
The logical continuation of Paper IV is an in vivo therapy study investigating co-treatment with AT13387 and radiation to determine an optimal
dose and time schedules to potentiate the combination effect. In such a
study, radiation treatment could be administered as an external beam of radi51
ation or in the form of radio-immunotherapy. For this purpose, CD44v6
targeting is promising, because the expression of this molecule is not affected by the inhibition of HSP90. CD44v6 expression has been associated with
increased radioresistance, and the targeting of CD44v6 high-expressing cells
is of particular interest.
Paper V demonstrated that monitoring of EGFR with PET/CT is a novel
approach to investigate the treatment response to AT13387. Other EGFR
tracers, preferably smaller conjugates could be tested that allow imaging at
earlier time points with high tumor uptake rates and improved contrast. Additionally, imaging at different time points could be beneficial.
Another appealing approach is to monitor the response of the constitutively auto-phosphorylated EGFR variant EGFRvIII in combination with the
inhibition of HSP90.
52
Acknowledgements
I would like to thank everyone who has contributed to this thesis.
In particular, I would like to thank the following people:
Marika Nestor, my supervisor and friend, for your creative ideas, support
and inspiring discussions about science, family and life. I appreciate that you
always have had time to answer my questions and solve problems, even
when you were extremely busy. I truly admire your efficiency and devotion
to your work. Your optimism and passion for science is an inspiration!
My co-supervisor, Bo Stenerlöw, for welcoming me into your group without
any prior knowledge of me. You have taught me a lot about science, and
your encouragement has meant a lot to me. Thank you for your wise guidance, caring, patience, kindness and fairness during these years.
My co-supervisor, Johan Lennartsson, for making this thesis possible and
for introducing me to the people at the Ludwig Institute of Cancer Research.
I would like to thank all my collaborators and co-authors. Without you the
articles in this thesis would not have been possible, especially the following
people:
Johan Nilvebrant for a great job with the anti-CD44v6 targeting fragments.
Karl Sandström and Anna-Karin Haylock for help with the animal experiments and for showing me how to hold a scalpel and scissors “like a real
surgeon”.
Olof Eriksson and Ram Kumar Selvaraju for all of the discussions and invaluable assistance with the PET analysis.
Jan Siljason for help with immunohistochemistry and the analysis.
53
All of the people who belong in one or the other way to the “former BMS”
and Ridgeview crew. Thanks for all our enjoyable discussions at fika and
over lunch, the many great parties, funny movies and for making BMS a
great place to belong. Because of you, it is fun to go to work every day!
Especially to:
Jörgen for recruiting me to the BMS team.
Vladimir for advice, discussions and help with the experimental setup.
Kicki for all of your care, cheerful spirit and love. You have given me the
feeling that I have a family here in Sweden.
Helene for your valuable help with administration and that I could always
talk to you even when it was not so easy for me.
Kalle, Hanna, John and Jos for interesting ideas and help with the LigandTracer measurements.
Lina and Lovisa for introduction into the obscure world of Western blotting.
Sara S, my supervisor, when I started as a Master’s student. Thank you for
inspiring discussions and for teaching me ~100 lab techniques in 2 weeks.
Ann-Sofie for enjoyable discussions about dogs, cats, horses and children.
Amelie for being such an inspiring person; your “can do” attitude sets a high
benchmark for us all.
Sara A, Mohamed, Dan, Eva, Joanna and Javad for your kindness, friendly
chats in the corridor and for always helping me when I needed assistance.
Andris, thank you for listening to all of my complaints, for climbing and
“fooding”, and for all of the scientific and silly debates.
Pavel, thank you for all of the interesting discussions, for always caring and
for great company at several conferences. I love going sightseeing with you!
Anja and Jonas, you are a real energy boost for our group! Thank you for
your support, babysitting and great conference company. Your positive attitude is extremely contagious!
54
My dear friend, Hadis, for going through up’s and down’s over all of these
years with me and for the emergency chocolate in your desk drawer. Your
smile lightens a gloomy day.
Thank you to my colleagues at the Preclinical Pet Platform, Anna, Olof,
Veronika, Sergio, Irina, Ram, Marie, Maria, Jennie, Zohreh and Bogdan.
I would like to thank “my” project students: Ram, Gamze, Adrian, Agnes
and Frida for all the hours you spent in the lab and analyzing data. This
thesis is your work, too! I think I have learned as much from you as you
from me.
Thank you to the UGSBR team for the lovely get-togethers: Hannah, Charlotte, Johan, Anne-Li, Theodora, Xiang and Jennie.
To all of my friends in Germany, Sweden and in all other places worldwide,
thank you for showing me that there is a life outside the lab! Thank you for
discussions, Skype chats, conventions, cooking, baking, watching movies,
board games (and letting me win), role playing, running, exercising, ice skating, watching hockey games, climbing, boxing, playing cards, paint ball,
badminton, kubb and brännboll, BBQing, shooting, sword fighting, picking
berries or mushrooms, traveling, hiking, sumo wrestling or just hanging out.
Vielen lieben Dank an meine Familie in Deutschland, für all eure Hilfe und
Unterstützung, für unvergessliche Urlaube, Ausflüge, gutes Essen und
unzählige Skype-Telefonate.
Amelie und Klein-L. Danke, dass es euch gibt und dass ich euch auf eurem
Weg begleiten darf.
Christer, my love and my best friend. Thank you for your incredible patience, love and support, and for always making me smile. You are the best
thing that ever happened to me ♥.
55
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
56
Jemal, A., et al., Global cancer statistics. CA Cancer J Clin, 2011. 61(2): p.
69-90.
Siegel, R., D. Naishadham, and A. Jemal, Cancer statistics, 2012. CA
Cancer J Clin, 2012. 62(1): p. 10-29.
Siegel, R., et al., Cancer treatment and survivorship statistics, 2012. CA
Cancer J Clin, 2012. 62(4): p. 220-41.
Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation.
Cell, 2011. 144(5): p. 646-74.
Weinberg, R., How cancer arises. Scientific American, 1996. 275(3): p.
62-70.
Ian F. Tannock, L.H., Robert G. Bristow, The basic science of oncology,
4th edition. 2005, New Baskerville: McGraw-Hill Companies.
Hanahan, D. and R.A. Weinberg, The hallmarks of cancer. Cell, 2000.
100(1): p. 57-70.
Jordan, C.T., M.L. Guzman, and M. Noble, Cancer Stem Cells. N Engl J
Med, 2006. 355(12): p. 1253-1261.
Leemans, C.R., B.J. Braakhuis, and R.H. Brakenhoff, The molecular
biology of head and neck cancer. Nat Rev Cancer, 2011. 11(1): p. 9-22.
Belcher, R., et al., Current treatment of head and neck squamous cell
cancer. J Surg Oncol, 2014. 110(5): p. 551-74.
Patel, K., et al., Heat shock protein 90 (HSP90) is overexpressed in p16negative oropharyngeal squamous cell carcinoma, and its inhibition in
vitro potentiates the effects of chemoradiation. Cancer Chemother
Pharmacol, 2014. 74(5): p. 1015-22.
Spiegelberg, D., et al., Characterization of CD44 variant expression in
head and neck squamous cell carcinomas. Tumor Biol, 2014. 35(3): p.
2053-62.
de Castro-Carpeno, J., et al., EGFR and colon cancer: a clinical view. Clin
Transl Oncol, 2008. 10(1): p. 6-13.
Ng, K. and A.X. Zhu, Targeting the epidermal growth factor receptor in
metastatic colorectal cancer. Crit Rev Oncol Hematol, 2008. 65(1): p. 820.
Huxley, R.R., et al., The impact of dietary and lifestyle risk factors on risk
of colorectal cancer: a quantitative overview of the epidemiological
evidence. Int J Cancer, 2009. 125(1): p. 171-80.
Loupakis, F., et al., PTEN expression and KRAS mutations on primary
tumors and metastases in the prediction of benefit from cetuximab plus
irinotecan for patients with metastatic colorectal cancer. J Clin Oncol,
2009. 27(16): p. 2622-9.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
Hynes, N.E. and H.A. Lane, ERBB receptors and cancer: the complexity of
targeted inhibitors. Nat Rev Cancer, 2005. 5(5): p. 341-54.
Heinemann, V., et al., Clinical relevance of EGFR- and KRAS-status in
colorectal cancer patients treated with monoclonal antibodies directed
against the EGFR. Cancer Treat Rev, 2009. 35(3): p. 262-71.
Todaro, M., et al., CD44v6 is a marker of constitutive and reprogrammed
cancer stem cells driving colon cancer metastasis. Cell stem cell, 2014.
14(3): p. 342-356.
Weissleder, R., Molecular imaging in cancer. Science, 2006. 312(5777): p.
1168-71.
Bruce Alberts, A.J., Julian Lewis, Martin Raff, Keith Roberts, and Peter
Walter, Molecular Biology of the Cell, 4th edition. 2002, New York:
Garland Science.
Mladenov, E., et al., DNA double-strand break repair as determinant of
cellular radiosensitivity to killing and target in radiation therapy. Front
Oncol, 2013. 3: p. 113.
Eriksson, D. and T. Stigbrand, Radiation-induced cell death mechanisms.
Tumor Biol, 2010. 31(4): p. 363-372.
De Palma, M. and D. Hanahan, The biology of personalized cancer
medicine: facing individual complexities underlying hallmark capabilities.
Mol Oncol, 2012. 6(2): p. 111-27.
Compton, C., Getting to personalized cancer medicine: taking out the
garbage. Cancer, 2007. 110(8): p. 1641-3.
Mammatas, L., et al., Molecular imaging of targeted therapies with
positron emission tomography: the visualization of personalized cancer
care. Cell Oncol (Dordr)., 2014: p. 1-16.
Parkinson, D.R., B.E. Johnson, and G.W. Sledge, Making personalized
cancer medicine a reality: challenges and opportunities in the development
of biomarkers and companion diagnostics. Clin Cancer Res, 2012. 18(3): p.
619-24.
Reilly, R., Aiming for a direct hit: combining molecular imaging with
targeted cancer therapy. J Nucl Med, 2009. 50(7): p. 1017-1019.
James, M.L. and S.S. Gambhir, A Molecular Imaging Primer: Modalities,
Imaging Agents, and Applications. Vol. 92. 2012. 897-965.
Corcoran, E.B. and R.N. Hanson, Imaging EGFR and HER2 by PET and
SPECT: a review. Med Res Rev, 2014. 34(3): p. 596-643.
Madsen, M.T., Recent advances in SPECT imaging. J Nucl Med, 2007.
48(4): p. 661-73.
Zhang, Y., H. Hong, and W. Cai, PET tracers based on Zirconium-89. Curr
Radiopharm, 2011. 4(2): p. 131-9.
Nayak, T.K. and M.W. Brechbiel, Radioimmunoimaging with longer-lived
positron-emitting radionuclides: potentials and challenges. Bioconjug
Chem, 2009. 20(5): p. 825-41.
Lee, F.T. and A.M. Scott, Immuno-PET for tumor targeting. J Nucl Med,
2003. 44(8): p. 1282-3.
Townsend, D.W., et al., PET/CT today and tomorrow. J Nucl Med, 2004.
45 Suppl 1: p. 4S-14S.
Boross, P. and J.H. Leusen, Mechanisms of action of CD20 antibodies. Am
J Cancer Res, 2012. 2(6): p. 676-90.
57
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
58
Salaun, P.Y., et al., Phase II trial of anticarcinoembryonic antigen
pretargeted radioimmunotherapy in progressive metastatic medullary
thyroid carcinoma: biomarker response and survival improvement. J Nucl
Med, 2012. 53(8): p. 1185-92.
DeNardo, G.L., S.J. DeNardo, and R. Balhorn, Systemic radiotherapy can
cure lymphoma: a paradigm for other malignancies? Cancer Biother
Radiopharm, 2008. 23(4): p. 383-97.
Eriksson, S.E., et al., Sequential radioimmunotherapy with 177Lu- and
211At-labeled monoclonal antibody BR96 in a syngeneic rat colon
carcinoma model. Cancer Biother Radiopharm, 2014. 29(6): p. 238-46.
Jain, M., et al., Emerging trends for radioimmunotherapy in solid tumors.
Cancer Biother Radiopharm, 2013. 28(9): p. 639-50.
Chamarthy, M.R. and S.C. Williams, Radioimmunotherapy of nonHodgkin's lymphoma: from the 'magic bullets' to 'radioactive magic
bullets'. Yale J Biol Med, 2011. 84(4): p. 391–407.
Sofou, S., Radionuclide carriers for targeting of cancer. Int J
Nanomedicine, 2008. 3(2): p. 181-99.
Kelkar, S.S. and T.M. Reineke, Theranostics: combining imaging and
therapy. Bioconjug Chem, 2011. 22(10): p. 1879-903.
Baum, R.P. and H.R. Kulkarni, THERANOSTICS: From Molecular
Imaging Using Ga-68 Labeled Tracers and PET/CT to Personalized
Radionuclide Therapy - The Bad Berka Experience. Theranostics, 2012.
2(5): p. 437-47.
Perez, A., et al., CD44 interacts with EGFR and promotes head and neck
squamous cell carcinoma initiation and progression. Oral Oncol, 2013.
49(4): p. 306-13.
Marhaba, R. and M. Zöller, CD44 in Cancer Progression: Adhesion,
Migration and Growth Regulation. J Mol Histol, 2004. 35(3): p. 211-231.
Prince, M.E., et al., Identification of a subpopulation of cells with cancer
stem cell properties in head and neck squamous cell carcinoma. Proc Natl
Acad Sci U S A, 2007. 104(3): p. 973-8.
Wielenga, V.J.M., et al., Expression of CD44 Variant Proteins in Human
Colorectal Cancer Is Related to Tumor Progression. Cancer Res, 1993.
53(20): p. 4754-4756.
Sahlberg, S.H., et al., Evaluation of cancer stem cell markers CD133,
CD44, CD24: association with AKT isoforms and radiation resistance in
colon cancer cells. PLoS One, 2014. 9(4): p. e94621.
Bhaijee, F., et al., Cancer stem cells in head and neck squamous cell
carcinoma: a review of current knowledge and future applications. Head
Neck, 2012. 34(6): p. 894-9.
Orian-Rousseau, V. and H. Ponta, Perspectives of CD44 targeting
therapies. Arch Toxicol, 2015. 89(1): p. 3-14.
Uniprot, C. http://www.uniprot.org/uniprot/P16070. [cited 2015 March].
Orian-Rousseau, V., CD44, a therapeutic target for metastasising tumours.
Eur J Cancer, 2010. 46(7): p. 1271-7.
Heider, K.-H., et al., CD44v6: a target for antibody-based cancer therapy.
Cancer Immunol Immunother, 2004. 53(7): p. 567-579.
Normanno, N., et al., Epidermal growth factor receptor (EGFR) signaling
in cancer. Gene, 2006. 366(1): p. 2-16.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
Ciardiello, F. and G. Tortora, EGFR antagonists in cancer treatment. N
Engl J Med, 2008. 358(11): p. 1160-74.
Mishani, E., et al., Imaging of EGFR and EGFR tyrosine kinase
overexpression in tumors by nuclear medicine modalities. Curr Pharm Des,
2008. 14(28): p. 2983-98.
Yarden, Y. and M.X. Sliwkowski, Untangling the ErbB signalling network.
Nat Rev Mol Cell Biol, 2001. 2(2): p. 127-37.
Hong, D., et al., Targeting the molecular chaperone heat shock protein 90
(HSP90): lessons learned and future directions. Cancer Treat Rev, 2013.
39(4): p. 375-387.
Den, R. and B. Lu, Heat shock protein 90 inhibition: rationale and clinical
potential. Ther Adv Med Oncol, 2012. 4(4): p. 211-218.
Subbarao Sreedhar, A., et al., Hsp90 isoforms: functions, expression and
clinical importance. FEBS Letters, 2004. 562(1-3): p. 11-15.
Jhaveri, K., et al., Heat shock protein 90 inhibitors in the treatment of
cancer: current status and future directions. Expert Opin Investig Drugs,
2014. 23(5): p. 611-28.
Lu, X., et al., Hsp90 inhibitors and drug resistance in cancer: the potential
benefits of combination therapies of Hsp90 inhibitors and other anticancer drugs. Biochem Pharmacol, 2012. 83(8): p. 995-1004.
Porter, J.R., C.C. Fritz, and K.M. Depew, Discovery and development of
Hsp90 inhibitors: a promising pathway for cancer therapy. Curr Opin
Chem Biol, 2010. 14(3): p. 412-20.
Tsutsumi, S., K. Beebe, and L. Neckers, Impact of heat-shock protein 90
on cancer metastasis. Future Oncol (London, England), 2009. 5(5): p. 679688.
Koga, F., K. Kihara, and L. Neckers, Inhibition of cancer invasion and
metastasis by targeting the molecular chaperone heat-shock protein 90.
Anticancer res, 2009. 29(3): p. 797-807.
Ahsan, A., et al., Wild-type EGFR is stabilized by direct interaction with
HSP90 in cancer cells and tumors. Neoplasia (New York, N.Y.), 2012.
14(8): p. 670-677.
Falsone, S., et al., A proteomic snapshot of the human heat shock protein
90 interactome. FEBS lett, 2005. 579(28): p. 6350-6354.
Lavictoire, S., et al., Interaction of Hsp90 with the nascent form of the
mutant epidermal growth factor receptor EGFRvIII. J Biol Chem, 2003.
278(7): p. 5292-5299.
Shimamura, T., et al., Hsp90 inhibition suppresses mutant EGFR-T790M
signaling and overcomes kinase inhibitor resistance. Cancer Res, 2008.
68(14): p. 5827-5838.
Dickson, M. and J.P. Gagnon, Key factors in the rising cost of new drug
discovery and development. Nat Rev Drug Discov, 2004. 3(5): p. 417-29.
Begley, C.G. and L.M. Ellis, Drug development: Raise standards for
preclinical cancer research. Nature, 2012. 483(7391): p. 531-3.
Scott, A.M., J.D. Wolchok, and L.J. Old, Antibody therapy of cancer. Nat
Rev Cancer, 2012. 12(4): p. 278-87.
Wu, A.M., Engineered antibodies for molecular imaging of cancer.
Methods, 2014. 65(1): p. 139-47.
59
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
60
Romer, T., H. Leonhardt, and U. Rothbauer, Engineering antibodies and
proteins for molecular in vivo imaging. Curr Opin Biotechnol, 2011. 22(6):
p. 882-7.
Sharkey, R.M. and D.M. Goldenberg, Targeted therapy of cancer: new
prospects for antibodies and immunoconjugates. CA Cancer J Clin, 2006.
56(4): p. 226-43.
Ståhl, S., et al., Affinity proteins and their generation. J Chem Technol
Biotechnol, 2013. 88(1): p. 25-38.
Stroomer, J.W., et al., Safety and biodistribution of 99mTechnetium-labeled
anti-CD44v6 monoclonal antibody BIWA 1 in head and neck cancer
patients. Clin Cancer Res, 2000. 6(8): p. 3046-55.
Postema, E.J., et al., Dosimetric analysis of radioimmunotherapy with
186Re-labeled bivatuzumab in patients with head and neck cancer. J Nucl
Med, 2003. 44(10): p. 1690-9.
Van Hal, N.L., et al., Sequence variation in the monoclonal-antibody-U36defined CD44v6 epitope. Cancer Immunol Immunother, 1997. 45(2): p. 8892.
Colnot, D., et al., Safety, biodistribution, pharmacokinetics, and
immunogenicity of 99mTc-labeled humanized monoclonal antibody BIWA
4 (bivatuzumab) in patients with squamous cell carcinoma of the head and
neck. Cancer Immunol Immunother, 2003. 52(9): p. 576-582.
Tijink, B.M., et al., A phase I dose escalation study with anti-CD44v6
bivatuzumab mertansine in patients with incurable squamous cell
carcinoma of the head and neck or esophagus. Clin Cancer Res, 2006.
12(20 Pt 1): p. 6064-72.
Riechelmann, H., et al., Phase I trial with the CD44v6-targeting
immunoconjugate bivatuzumab mertansine in head and neck squamous cell
carcinoma. Oral Oncol, 2008. 44(9): p. 823-9.
Nestor, M.V., Targeted radionuclide therapy in head and neck cancer.
Head & neck, 2010. 32(5): p. 666-678.
Colnot, D.R., et al., Reinfusion of unprocessed, granulocyte colonystimulating factor-stimulated whole blood allows dose escalation of
186Relabeled chimeric monoclonal antibody U36 radioimmunotherapy in
a phase I dose escalation study. Clin Cancer Res, 2002. 8(11): p. 3401-6.
Borjesson, P.K., et al., Radiation dosimetry of 89Zr-labeled chimeric
monoclonal antibody U36 as used for immuno-PET in head and neck
cancer patients. J Nucl Med, 2009. 50(11): p. 1828-36.
Sandstrom, K., et al., A novel CD44v6 targeting antibody fragment with
improved tumor-to-blood ratio. Int J Oncol, 2012. 40(5): p. 1525-32.
Nilvebrant, J., et al., Selection and in vitro characterization of human
CD44v6-binding antibody fragments. Biotechnol Appl Biochem, 2012.
59(5): p. 367-80.
Milenic, D.E., et al., Cetuximab: preclinical evaluation of a monoclonal
antibody
targeting
EGFR
for
radioimmunodiagnostic
and
radioimmunotherapeutic applications. Cancer Biother Radiopharm, 2008.
23(5): p. 619-31.
Nayak, T., et al., PET imaging of HER1-expressing xenografts in mice with
86Y-CHX-A″-DTPA-cetuximab. Eur J Nucl Med Mol Imaging, 2010. 37(7):
p. 1368-1376.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
Perk, L.R., et al., 89Zr as a PET Surrogate Radioisotope for Scouting
Biodistribution of the Therapeutic Radiometals 90Y and 177Lu in TumorBearing Nude Mice After Coupling to the Internalizing Antibody
Cetuximab. J Nucl Med, 2005. 46(11): p. 1898-1906.
Cai, W., et al., Quantitative PET of EGFR expression in xenograft-bearing
mice using 64Cu-labeled cetuximab, a chimeric anti-EGFR monoclonal
antibody. Eur J Nucl Med Mol Imaging, 2007. 34(6): p. 850-8.
van Dijk, L.K., et al., 111In-Cetuximab-F(ab')2 SPECT and 18F-FDG PET
for Prediction and Response Monitoring of Combined-Modality Treatment
of Human Head and Neck Carcinomas in a Mouse Model. J Nucl Med,
2015. 56(2): p. 287-92.
Sandstrom, K., et al., Improved tumor-to-organ ratios of a novel 67Gahuman epidermal growth factor radionuclide conjugate with
preadministered antiepidermal growth factor receptor affibody molecules.
Cancer Biother Radiopharm, 2011. 26(5): p. 593-601.
Capala, J., et al., Radiolabeling of epidermal growth factor with 99mTc and
in vivo localization following intracerebral injection into normal and
glioma-bearing rats. Bioconjug Chem, 1997. 8(3): p. 289-95.
Cornelissen, B., et al., ErbB-2 blockade and prenyltransferase inhibition
alter epidermal growth factor and epidermal growth factor receptor
trafficking and enhance (111)In-DTPA-hEGF Auger electron radiation
therapy. J Nucl Med, 2011. 52(5): p. 776-83.
Velikyan, I., Å. Sundberg, and Ö. Lindhe, Preparation and evaluation of
68Ga-DOTA-hEGF for visualization of EGFR expression in malignant
tumors. J Nucl Med, 2005. 46(11): p. 1881-1888.
Li, W., et al., PET imaging of EGF receptors using [18F]FBEM-EGF in a
head and neck squamous cell carcinoma model. Eur J Nucl Med Mol
Imaging, 2012. 39(2): p. 300-8.
Orlova, A., et al., Update: affibody molecules for molecular imaging and
therapy for cancer. Cancer Biother Radiopharm, 2007. 22(5): p. 573-84.
Tolmachev, V., et al., Imaging of EGFR expression in murine xenografts
using site-specifically labelled anti-EGFR 111In-DOTA-ZEGFR: 2377
Affibody molecule: aspect of the injected tracer amount. Eur J Nucl Med
Mol Imaging, 2010. 37(3): p. 613-622.
Gan, H.K., et al., Targeting of a conformationally exposed, tumor-specific
epitope of EGFR as a strategy for cancer therapy. Cancer Res, 2012.
72(12): p. 2924-30.
Sharp, S. and P. Workman, Inhibitors of the HSP90 molecular chaperone:
current status. Adv Cancer Res, 2006. 95: p. 323-48.
Roe, S.M., et al., Structural basis for inhibition of the Hsp90 molecular
chaperone by the antitumor antibiotics radicicol and geldanamycin. J Med
Chem, 1999. 42(2): p. 260-6.
Vanneman, M. and G. Dranoff, Combining immunotherapy and targeted
therapies in cancer treatment. Nat Rev Cancer, 2012. 12(4): p. 237-51.
Kabakov, A.E., V.A. Kudryavtsev, and V.L. Gabai, Hsp90 inhibitors as
promising agents for radiotherapy. J Mol Med (Berl), 2010. 88(3): p. 2417.
61
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
62
Hong, D.S., et al., Targeting the molecular chaperone heat shock protein
90 (HSP90): lessons learned and future directions. Cancer Treat Rev,
2013. 39(4): p. 375-87.
Kim, T., G. Keum, and A.N. Pae, Discovery and development of heat shock
protein 90 inhibitors as anticancer agents: a review of patented potent
geldanamycin derivatives. Expert Opin Ther Pat, 2013. 23(8): p. 919-43.
Niewidok, N., et al., Hsp90 Inhibitors NVP-AUY922 and NVP-BEP800 May
Exert a Significant Radiosensitization on Tumor Cells along with a Cell
Type-Specific Cytotoxicity. Transl Oncol, 2012. 5(5): p. 356-369.
Kamal, A., et al., A high-affinity conformation of Hsp90 confers tumour
selectivity on Hsp90 inhibitors. Nature, 2003. 425(6956): p. 407-10.
Samuni, Y., et al., Reactive oxygen species mediate hepatotoxicity induced
by the Hsp90 inhibitor geldanamycin and its analogs. Free Radic Biol
Med, 2010. 48(11): p. 1559-63.
Woodhead, A.J., et al., Discovery of (2,4-dihydroxy-5-isopropylphenyl)-[5(4-methylpiperazin-1-ylmethyl)-1,3-dihydrois
oindol-2-yl]methanone
(AT13387), a novel inhibitor of the molecular chaperone Hsp90 by
fragment based drug design. J Med Chem, 2010. 53(16): p. 5956-69.
Kang, M.H., et al., Initial testing (Stage 1) of AT13387, an HSP90
inhibitor, by the pediatric preclinical testing program. Pediatr Blood
Cancer, 2012. 59(1): p. 185-8.
Shapiro, G.I., et al., First-in-human phase I dose escalation study of a
second-generation non-ansamycin HSP90 inhibitor, AT13387, in patients
with advanced solid tumors. Clin Cancer Res, 2015. 21(1): p. 87-97.
Jhaveri, K., et al., Advances in the clinical development of heat shock
protein 90 (Hsp90) inhibitors in cancers. Biochim Biophys Acta, 2012.
1823(3): p. 742-55.
Koll, T.T., et al., HSP90 inhibitor, DMAG, synergizes with radiation of
lung cancer cells by interfering with base excision and ATM-mediated
DNA repair. Mol Cancer Ther, 2008. 7(7): p. 1985-92.
Matsumoto, Y., H. Machida, and N. Kubota, Preferential sensitization of
tumor cells to radiation by heat shock protein 90 inhibitor geldanamycin. J
Radiat Res, 2005. 46(2): p. 215-21.
Gandhi, N., et al., Novel Hsp90 inhibitor NVP-AUY922 radiosensitizes
prostate cancer cells. Cancer Biol Ther, 2013. 14(4): p. 347-56.
Stingl, L., et al., Novel HSP90 inhibitors, NVP-AUY922 and NVP-BEP800,
radiosensitise tumour cells through cell-cycle impairment, increased DNA
damage and repair protraction. Br J Cancer, 2010. 102(11): p. 1578-91.
Machida, H., et al., Geldanamycin, an inhibitor of Hsp90, sensitizes human
tumour cells to radiation. Int J Radiat Biol, 2003. 79(12): p. 973-80.
Nestor, M., K. Andersson, and H. Lundqvist, Characterization of 111In
and 177Lu-labeled antibodies binding to CD44v6 using a novel automated
radioimmunoassay. J Mol Recognit, 2008. 21(3): p. 179-83.
Stenberg, J., et al., Choice of labeling and cell line influences interactions
between the Fab fragment AbD15179 and its target antigen CD44v6. Nucl
Med Biol., 2014. 41(2): p. 140-147.
Acta Universitatis Upsaliensis
Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1085
Editor: The Dean of the Faculty of Medicine
A doctoral dissertation from the Faculty of Medicine, Uppsala
University, is usually a summary of a number of papers. A few
copies of the complete dissertation are kept at major Swedish
research libraries, while the summary alone is distributed
internationally through the series Digital Comprehensive
Summaries of Uppsala Dissertations from the Faculty of
Medicine. (Prior to January, 2005, the series was published
under the title “Comprehensive Summaries of Uppsala
Dissertations from the Faculty of Medicine”.)
Distribution: publications.uu.se
urn:nbn:se:uu:diva-247539
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2015